WO1995005465A1 - Erythropoietin analogs - Google Patents

Erythropoietin analogs Download PDF

Info

Publication number
WO1995005465A1
WO1995005465A1 PCT/US1994/009257 US9409257W WO9505465A1 WO 1995005465 A1 WO1995005465 A1 WO 1995005465A1 US 9409257 W US9409257 W US 9409257W WO 9505465 A1 WO9505465 A1 WO 9505465A1
Authority
WO
WIPO (PCT)
Prior art keywords
thr
asn
epo
erythropoietin
ser
Prior art date
Application number
PCT/US1994/009257
Other languages
French (fr)
Inventor
Steven G. Elliott
Thomas E. Byrne
Original Assignee
Amgen Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22319789&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1995005465(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to AU76327/94A priority Critical patent/AU677097B2/en
Priority to UA95058432A priority patent/UA49793C2/en
Priority to HU9501435A priority patent/HU220334B/en
Priority to JP7507153A priority patent/JP2938572B2/en
Priority to SK502-95A priority patent/SK282003B6/en
Priority to KR1019950701453A priority patent/KR100328769B1/en
Application filed by Amgen Inc. filed Critical Amgen Inc.
Priority to NZ273134A priority patent/NZ273134A/en
Priority to CA002147124A priority patent/CA2147124C/en
Publication of WO1995005465A1 publication Critical patent/WO1995005465A1/en
Priority to NO19951445A priority patent/NO323104B1/en
Priority to FI951792A priority patent/FI117136B/en
Priority to KR1019957001453A priority patent/KR960701994A/en
Priority to LVP-95-99A priority patent/LV10972B/en
Priority to NO2007008C priority patent/NO2007008I2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to erythropoietin analogs having at least one additional site for glycosylation or a rearrangement of at least one site for glycosylation.
  • the invention also relates to DNA sequences encoding said erythropoietin analogs, and recombinant plasmids and host cells for analog expression.
  • Erythropoietin is a glycoprotein hormone involved in the maturation of erythroid progenitor cells into erythrocytes. It is essential in regulating levels of red blood cells in circulation. Naturally occurring erythropoietin is produced by the liver during fetal life and by the kidney of adults and circulates in the blood and stimulates the production of red blood cells in bone marrow. Anemia is almost invariably a
  • glycosylation can dramatically affect the physical properties of proteins and can also be important in protein stability, secretion, and
  • Glycosylation occurs at specific locations along the polypeptide backbone and is usually of two types: O-linked oligosaccharides are attached to serine or threonine residues while N-linked oligosaccharides are attached to asparagine residues when they are part of the sequence Asn-X-Ser/Thr, where X can be any amino acid except proline.
  • the structures of N-linked and O-linked oligosaccharides and the sugar residues found in each type are different.
  • One type of sugar that is commonly found on both is N-acetylneuraminic acid
  • Sialic acid is usually the terminal residue of both N-linked and
  • O-linked oligosaccharides and, by virtue of its negative charge, may confer acidic properties to the
  • glycoprotein glycoprotein
  • Both human urinary derived erythropoietin and recombinant erythropoietin (expressed in mammalian cells) having the amino acid sequence 1-165 of human erythropoietin contain three N-linked and one O-linked oligosaccharide chains which together comprise about 40% of the total molecular weight of the glycoprotein.
  • N-linked glycosylation occurs at asparagine residues located at positions 24, 38 and 83 while O-linked glycosylation occurs at a serine residue located at position 126 (Lai et al. J. Biol. Chem. 261, 3116
  • Enzymatic treatment of glycosylated erythropoietin to remove all sialic acid residues results in a loss of in vivo activity but does not affect in vitro activity
  • Glycoproteins such as erythropoietin can be separated into different charged forms using techniques such as isoelectric focusing (IEF) .
  • IEF isoelectric focusing
  • erythropoietin having a defined sialic acid content and biological activity.
  • Pharmaceutical compositions containing such molecules would have therapeutic benefit.
  • glycosylation The added sites for glycosylation may result in a greater number of carbohydrate chains, and higher sialic acid content, than human erythropoietin.
  • Erythropoietin analogs comprising amino acid sequences which include the rearrangement of at least one site for glycosylation are also provided.
  • Analogs comprising an addition of one or more amino acids to the carboxy terminal end of erythropoietin wherein the addition provides at least one glycosylation site are also included.
  • the invention further encompasses DNA
  • Figure 1 shows an analytical isoelectric focusing gel of the separate recombinant erythropoietin isoforms.
  • Gel lanes 1-11 show isoforms ranging from less acidic (higher pi) in lane 1 to more acidic (lower pI), in lane 11.
  • Purified recombinant erythropoietin containing a mixture of isoforms 9-14 is also shown in the far left and right lanes of the gel.
  • Figure 2 shows the relationship between the number of sialic acids per erythropoietin isoform and the in vivo specific activity of each isoform expressed as units per mg of erythropoietin polypeptide.
  • Figure 3 shows an analytical isoelectric focusing gel of defined mixtures of recombinant
  • erythropoietin isoforms prepared by anion exchange chromatography under different conditions.
  • Gel lanes 1-6 represent, respectively, erythropoietin isoforms eluted in a high salt wash after washing the Q-Sepharose fast flow column with 150 mM acetic acid, pH 4.7, 150 mM acetic acid (unbuffered), 200 mM acetic acid, pH 4.7, 250 mM acetic acid, pH 4.7, 300 mM acetic acid, pH 4.7 or 300 mM acetic acid (unbuffered).
  • 150 mM acetic acid pH 4.7, 150 mM acetic acid (unbuffered)
  • 200 mM acetic acid pH 4.7, 250 mM acetic acid, pH 4.7, 300 mM acetic acid, pH 4.7 or 300 mM acetic acid (unbuffered).
  • erythropoietin isoforms 8 to 12 achieved by subjecting cell conditioned medium applied to a column of
  • Figure 5 shows the amino acid sequence of human erythropoietin. Squares indicate asparagine residues to which N-linked carbohydrate chains are attached and an asterisk indicates the serine residue modified with an O-linked carbohydrate chain.
  • Figures 6A, 6B, and 6C show the series of cloning steps used in generating plasmids for the construction and analysis of analogs of human
  • Figure 7 shows a Western blot analysis of COS cell supernatants of human sequence erythropoietin and indicated erythropoietin analogs.
  • the analogs [Asn 9 , Serli] EPO, [Asn 69 ] EPO, [Asn 125 , Ser 127 ] EPO, and
  • Figure 8 shows a Western blot analysis of COS cell supernatants of human sequence erythropoietin and indicated erythropoietin analogs after treatment with N-glycanase.
  • Thr 125 ] EPO were constructed as described in Example 6. The analogs [Val 126 ] EPO, [Pro 124 ] EPO, [Pro 125 ] EPO, [Thr 127 ] EPO, [Pro 125 , Ser 127 ] EPO and [Thr 125 , Ser 127 ] EPO are shown for comparison.
  • Figure 9 shows an isoelectric focusing gel of pools 2, 3 and 4 obtained by Q-Sepharose and C4 reverse phase chromatography of cell medium that supported the growth of CHO cells transfected with erythropoietin cDNA containing the [Thr 125 ] mutation.
  • Purified recombinant erythropoietin containing a mixture of isoforms were obtained using procedures described in Example 2 of Lai et al., supra, except that DEAE-Agarose chromatography is replaced by Q-Sepharose chromatography, is also shown in the left and right lanes of the gel.
  • Figure 10 shows a Western blot analysis of COS cell supernatants of recombinant human erythropoietin (rHuEPO) and selected analogs. The construction of the analogs is described in Example 6. Analogs N9, N14, N18, N19, N21, N24 and N39 have at least one additional carbohydrate chain as evidenced by slower gel mobility.
  • Figure 11 shows a Western blot analysis of COS cell supernatants of recombinant human erythropoietin and EPO N14 analog during N-glycanase digestion. Time points were taken at 0, 4, 12 and 45 minutes and after overnight digestion.
  • Figure 12 shows an isoelectric focusing gel of
  • EPO N14 analog isoform preparations The low isoform pool contains EPO N14 analog having mostly 6-12 sialic acids per molecule, the medium isoform pool contains analog N14 having mostly 10-15 sialic acids per
  • the high isoform pool contains EPO N14 analog having mostly 12-17 sialic acids per molecule.
  • Figure 13 shows the pharmacokinetics of recombinant human erythropoietin, isolated isoform 14 and EPO N14 analog (isoforms 15-17) after intravenous injection into rats.
  • Figure 14 shows a cold displacement assay of 125 I labeled recombinant human erythropoietin binding to the erythropoietin receptor in the presence of varying amounts of unlabeled rHuEPO, isolated isoform 14 or EPO N14 analog.
  • Figure 15 shows a mouse hematocrit study comparing the activity of EPO N14 high isoform pool (0.036 and 0.0712 ⁇ g), isolated isoform 14 (0.036 and 0.0712 ⁇ g), EPO 177 low isoform pool (0.0712 ⁇ g) and rHuEPO (0.071 ⁇ g).
  • the subject invention provides erythropoietin isoforms.
  • the specific isoforms of erythropoietin obtained in accordance with the present invention, and their properties, may vary depending upon the source of the starting material.
  • the isoforms of urinary derived human erythropoietin are different than the isoforms of recombinant erythropoietin.
  • the invention relates to an erythropoietin isoform having a specific number (i.e. a fixed number greater than 0) of sialic acids per erythropoietin molecule, said number selected from the group consisting of 1-14.
  • a specific number i.e. a fixed number greater than 0
  • sialic acids per erythropoietin molecule said number selected from the group consisting of 1-14.
  • said number is 9, 10, 11, 12, 13, or 14.
  • said number is greater than 14, advantageously 16-23.
  • erythropoietin isoform refers to erythropoietin preparations having a single isoelectric point (pI), and having the same amino acid sequence.
  • erythropoietin includes naturally occurring erythropoietin, urinary derived human erythropoietin as well as nonnaturally occurring polypeptides having an amino acid sequence and glycosylation sufficiently duplicative of that of naturally occurring erythropoietin to allow possession of in vivo biological properties of causing bone marrow cells to increase production of
  • erythropoietin corresponds to erythropoietin molecules having from 1-14 sialic acids, and each isoform present in purified recombinant erythropoietin has an in vivo activity which is related to the number of sialic acids the isoform possesses.
  • erythropoietin is the product of the expression of an exogenous DNA sequence that has been transfected into a non-human eucaryotic host cell, that is, in a preferred embodiment the erythropoietin is "recombinant erythropoietin".
  • Recombinant erythropoietin is advantageously produced according to the procedures described in commonly owned Lin U.S. Patent 4,703,008 hereby incorporated by
  • Recombinant erythropoietin can be purified according to the general procedures described in
  • Erythropoietin purified according to Example 2 of Lai et al. supra contains predominantly six isoforms when analyzed by IEF.
  • Example 4 (This more acidic form, migrating at >14 sialic acids on an IEF gel may contain nonsialic acid negative charges as shown by the resistance of some of the charge to sialidase digestion). These isoforms differ from each other by sialic acid content. As shown in the Examples, this is demonstrated by isolating 10 of these isoforms by preparative IEF and determining the sialic acid content of five of them. Of the isoforms assayed for sialic acid content, it is found that the five isoforms contained either 9, 10, 11, 12 or 13 sialic acid residues.
  • Isoforms 5 through 11 There is a relationship between the relative in vivo specific activity of erythropoietin and number of sialic acid residues per erythropoietin molecule from the isoforms 5 through 11 (each isoform is designated herein by the number of sialic acids per erythropoietin molecule). Isoforms 11 through 14 have approximately the same relative in vivo specific activity. Isoforms 5-14 were assayed for in vivo activity by the exhypoxic polycythemic mouse bioassay and the amount of each isoform present is determined by Bradford protein assay, absorbance at 280 nm or by radioimmunoassay (RIA) for erythropoietin. RIA determinations (Egrie et al.
  • polypeptide the average specific activity of purified erythropoietin as determined by RIA, to give protein concentrations of isolated isoforms or isoform mixtures expressed as mg erythropoietin polypeptide/ml.
  • erythropoietin polypeptide/ml the average specific activity of purified erythropoietin as determined by RIA
  • the in vivo specific activities referred to herein are measurements of relative in vivo specific activities and are not measurements of absolute in vivo specific activities. For the purposes of this
  • the specific activities are used only to compare relative activities of isoforms that have been assayed using the same assay, using the same conditions including the same internal standard, the same type of animals, having the same analysis of the data used to calculate specific activity, the same assay for
  • compositions comprising two or more erythropoietin isoforms.
  • the compositions comprise a mixture of isoforms having greater than a predetermined number of sialic acids per erythropoietin molecule, e.g. greater than 11 sialic acids per erythropoietin molecule, or greater than 12 sialic acids per molecule, e . g . a mixture of isoforms 12, 13 and 14.
  • compositions comprise mixtures of isoforms having a predetermined number of sialic acids per erythropoietin molecule, e .g. less than 12, but greater than 8 sialic acids per molecule as in, for example, a mixture of isoforms 9, 10, and 11 .
  • the invention also provides for compositions of
  • erythropoietin isoforms wherein the relative amounts of the isoforms are the same or different .
  • a mixture of isoforms 9, 10 and 11 could have the isoforms present in a variety of ratios such as 1 : 1 : 1 , 2 : 3 : 1 or 20 : 20 : 1.
  • compositions comprise mixtures of less than four isoforms, for example a mixture of isoforms 11, 12, and 13, or a mixture of 12 and 14, or a mixture of 7 and 13.
  • this invention also provides methods of isolating selected erythropoietin isoforms
  • ion exchange chromatography and chromatofocusing involve application of either crude human erythropoietin (cell conditioned media) or
  • the protein it is preferable to apply the protein to the column at about pH 7 while for purified preparations the protein can be applied to the column at pH 7 down to about pH 4 . After washing the column with buffer at about pH 4 , those erythropoietin isoforms that remain bound on the ion exchange column are eluted by
  • the isoforms are eluted from the column by a gradient of decreasing pH or by washing the column with a high concentration of salt .
  • individual isoforms are isolated using ion exchange chromatography .
  • isoform 14 was isolated using ion exchange chromatography as described in Example 8.
  • analogs of human erythropoietin refers to erythropoietin with one or more changes in the amino acid sequence of human erythropoietin which result in an increase in the number of sites for sialic acid
  • Analogs are generated by site-directed mutagenesis having additions, deletions, or
  • Such analogs may have a greater number of carbohydrate chains than human erythropoietin .
  • the erythropoietin analogs of the present invention comprise an amino acid sequence which includes at least one additional site for glycosylation .
  • Analogs having levels of sialic acid greater than those found in human erythropoietin are generated by adding
  • glycosylation sites which do not perturb the secondary or tertiary conformation required for biological
  • erythropoietin has 1, 2 or 3 additional sites for N-glycosylation or O-glycosylation, resulting in the addition of 1, 2, or 3 additional N-linked or O-linked carbohydrate chains.
  • a leucine at position 69 is replaced by an asparagine to give the sequence Asn-Leu-Ser, which serves as a fourth site for
  • N-glycosylation Such a change can commonly provide up to four additional sialic acids per molecule.
  • Examples of changes that generate additional O-glycosylation sites are alanine at position 125 to threonine and alanines at positions 124 and 125 to proline and threonine, respectively.
  • Analogs may be constructed which have one or more additional N-linked and O-linked chains, for example, analogs NO1 and N02 described in Table 5.
  • the subject invention includes many other analogs of human erythropoietin having additional sites for glycosylation.
  • Analogs having increased levels of carbohydrate attachment at a glycosylation site are also encompassed by the invention. Such analogs usually involve the substitution of one or more amino acids which are in close proximity to an N-linked or O-linked site. As a result of these amino acid changes, a greater proportion of erythropoietin polypeptides will have a carbohydrate modification.
  • the glycosylation sites may be naturally occurring or generated by
  • analog N13 does not generate an additional carbohydrate chain even though a
  • glycosylation site was introduced at position 88.
  • analogs N14 and N18 which have serine and valine residues, respectfully, substituted at position 87, have an additional carbohydrate chain at position 88.
  • analogs which have one or more amino acids extending from the carboxy terminal end of erythropoietin and wherein the carboxy terminal extension provides at least one
  • an analog was constructed by fusing the carboxy-terminal 28 amino acids of human chorionic gonadotropin (HCG) to the arginine residue at position 166 of human
  • HCG carboxy-terminal fragment has four sites for O-glycosylation (Kessler et al., J. Biol. Chem. 254, 7907 (1979)).
  • Tables 3, 4 and 5 list erythropoietin analogs which have additional sites for N-linked and/or O-linked carbohydrate chains.
  • the analogs have the sequence Asn-X-Ser/Thr substituted at various positions in the human erythropoietin polypeptide chain to create
  • Table 6 lists those analogs which add at least one additional N-linked or one additional O-linked carbohydrate chain, or add additional N-linked and
  • the erythropoietin analogs of the present invention also encompass erythropoietin having an amino acid sequence which includes a rearrangement of at least one site for glycosylation.
  • a rearrangement of a glycosylation site as used herein refers to the deletion of one or more glycosylation sites in human
  • Analogs R1, R2 and R3 are examples of such rearrangements and were constructed by deletion of the N-linked sites at positions 24, 38 or 83, respectively, and addition of an N-linked site at position 88.
  • carbohydrate site rearrangements are possible and the resulting analogs may or may not have a greater number of glycosylation sites compared to human erythropoietin.
  • positions of carbohydrate chains in erythropoietin may be changed in order to generate useful analogs without significantly affecting the biological activity.
  • erythropoietin analogs can be the product of expression of an exogenous DNA sequence, i.e., produced through recombinant DNA technology, or can be synthesized products.
  • An exogenous DNA sequence comprises cDNA, genomic DNA or chemically synthesized DNA encoding an erythropoietin analog.
  • Recombinant DNA plasmids and eucaryotic host cells useful for the expression of said analogs are also provided.
  • Expression vectors include any vector which is capable of expressing cloned DNA sequences in a eucaryotic host cell, particularly those vectors used for expression in COS and CHO cells. Examples of such vectors include plasmids pEC and pDEC ⁇ described in Example 6 of the specification. The cultivation of COS and CHO host cells expressing erythropoietin analogs was carried out using procedures known to one skilled in the art.
  • Isolated isoforms and mixtures, of isoforms derived from erythropoietin analogs are obtained using the methods described above for preparing isoforms of human erythropoietin. These methods may include isoelectric focusing, ion exchange chromatography and chromatofocusing. Preferably, ion exchange
  • Another embodiment of the invention relates to mammalian (e.g., Chinese Hamster Ovary, CHO) host cells which preferentially synthesize isoforms of human erythropoietin or erythropoietin analogs having greater than a specific number of sialic acids per molecule, e.g. greater than 10 sialic acids per molecule.
  • mammalian e.g., Chinese Hamster Ovary, CHO
  • Erythropoietin molecules have N-linked and O-linked oligosaccharides structures which can limit the sialic acid content of the molecule. For example,
  • tetraantennary four-branched N-linked oligosaccharides most commonly provide four possible sites for sialic acid attachment while bi- and triantennary
  • oligosaccharide chains which can substitute for the tetraantennary form at asparagine-linked sites, commonly have at most only two or three sialic acids attached.
  • O-linked oligosaccharides commonly provide two sites for sialic acid attachment.
  • erythropoietin molecules can accommodate a total of 14 sialic acid residues provided all three N-linked oligosaccharides are
  • tetraantennary Mammalian cell cultures are screened for those cells that preferentially add tetraantennary chains to recombinant erythropoietin, thereby maximizing the number of sites for sialic acid attachment.
  • the N-linked oligosaccharides of urinary erythropoietin contain sialic acid in both an ⁇ 2,3 and an ⁇ 2,6 linkage to galactose (Takeuchi et al. J. Biol. Chem. 263, 3657(1988)).
  • sialic acid in the ⁇ 2,3 linkage is added to galactose on the mannose ⁇ 1, 6 branch and the sialic acid in the ⁇ 2,6 linkage i's added to the galactose on the mannose ⁇ 1,3 branch.
  • the enzymes that add these sialic acids ⁇ -galactoside ⁇ 2,3 sialyltransferase and ⁇ -galactoside ⁇ 2, 6
  • sialyltransferase are most efficient at adding sialic acid to the mannose ⁇ 1,6 and mannose ⁇ 1,3 branches respectively.
  • DHFR Dihydrofolate reductase
  • Chinese Hamster Ovary (CHO) cells are a commonly used host cell for the production of recombinant glycoproteins including recombinant erythropoietin.
  • recombinant erythropoietin produced in CHO cells lacks sialic acid in the 2,6 linkage to galactose (Sasaki et al. (1987), supra; Takeuchi et al. (1987), supra).
  • human erythropoietin or an erythropoietin analog is produced in CHO cells that are transfected with a functional ⁇ -galactoside ⁇ 2, 6 sialyltransferase gene to give incorporation of sialic acid in ⁇ 2,6 linkage to galactose.
  • the resulting isoforms will contain sialic acid having both ⁇ 2,3 and ⁇ 2,6 linkages to galactose. See Lee et al. J. Biol. Chem. 264, 13848 (1989), hereby incorporated by reference, for a
  • compositions comprising a therapeutically effective amount of a specific isoform or mixture of isoforms together with a suitable diluent, adjuvant and/or carrier useful in erythropoietin therapy.
  • compositions comprising a therapeutically effective amount of an erythropoietin analog together with a suitable diluent, adjuvant and/or carrier are also encompassed.
  • a "therapeutically effective amount” as used herein refers to that amount which provides therapeutic effect for a given condition and
  • the administration of isoforms of human erythropoietin or erythropoietin analogs is preferably by parenteral routes. The specific route chosen will depend upon the condition being treated.
  • the administration of isoforms of human erythropoietin or erythropoietin analogs is preferably done as part of a formulation containing a suitable carrier, such as human serum albumin, a suitable diluent, such as a buffered saline solution, and/or a suitable adjuvant.
  • a suitable carrier such as human serum albumin
  • a suitable diluent such as a buffered saline solution
  • the required dosage will be in amounts sufficient to raise the hematocrit of patients and will vary depending upon the severity of the condition being treated, the method of administration used and the like.
  • erythropoietin standard used in the in vivo bioassays employed in the Examples is a recombinant erythropoietin standard that was standardized against a partially purified urinary erythropoietin standard.
  • erythropoietin standard used in the in vivo bioassays employed in the Examples.
  • the in vivo specific activities are expressed in "units/ml", “units/mg” and units/A 280 " and not as
  • IU/ml "IU/mg” and IU/A 280 " because the erythropoietin standard employed has not been directly correlated to any existing international standard.
  • Example 1 Isolation of Recombinant Erythropoietin Isoforms
  • Recombinant erythropoietin is produced as described in Lin, supra.
  • Recombinant erythropoietin used as starting material for the first and third isoform isolations is purified according to the
  • the sixth isoform preparation used as its starting material a purified preparation of recombinant erythropoietin having from 4 to 13 sialic residues.
  • This material was purified as per Example 2 of Lai et al. except for a modification to the ion exchange column (elution of the recombinant erythropoietin with a sodium chloride gradient at pH 8.4 and omission of the acetic acid/urea wash) which results in retention of most of the isoforms present in the starting material.
  • the first preparation approximately 20 mg of recombinant erythropoietin in 6.8 ml of 20 mM sodium citrate/ 100 mM sodium chloride, pH 7.0 are applied to the gel and focused at 8 watts for approximately 16 hours. After isoelectric focusing, the isoform bands in the gel are visualized by a paper contact print of the gel bed.
  • the print is made and then fixed by soaking in 3 changes (approximately 10 minutes each, room temperature) of fixing solution (40% methanol/10% acetic acid/10% TCA/3.5% sulfosalicylic acid), subjected to one change (approximately 10 minutes) of 40% methanol/10% acetic acid (30-60°C), stained for 15 minutes at 60°C in 0.125% Coomassie Blue R-250/40% methanol/10% acetic acid, and then destained in 7.5% methanol/10% acetic acid in order to visualize the separated isoforms.
  • fixing solution 50% methanol/10% acetic acid/10% TCA/3.5% sulfosalicylic acid
  • the region of the granulated gel bed containing the isoforms ( ⁇ 50% of the resin) is removed, water is added ( ⁇ 16 ml), and the slurry is poured into a 5.5 ⁇ 24.5 inch tray and evaporated to -40 g net weight. This preparation is focused for a second time and a contact print of the gel bed is made as before. The portion of gel containing each of the six discernible isoforms is removed from the gel bed.
  • erythropoietin in 21.8 ml of distilled water is applied to the gel and focused at 2 watts for 25 minutes, 10 watts for 20 hours and 15 watts for 15 minutes. Protein bands corresponding to the individual isoforms are observed visually and removed from the gel bed. Distilled water is added to gel-isolated isoforms to generate a slurry and the resulting supernatants are analyzed by analytical isoelectric focusing. An equal volume of 1 M Tris-HCl, pH 7.2 is added to each slurry, the suspensions are placed into separate small columns, and the liquid phase is allowed to flow through the column to elute the isoforms. Each flow through is concentrated and buffer exchanged to 20 mM sodium citrate/ 100 mM sodium chloride, pH 7.0 using Amicon disposable ultrafiltration devices having a 10,000 dalton molecular weight cutoff. An analytical
  • isoelectric focusing gel revealed that pools containing predominantly the single isoforms 9, 10, 11, 12, 13 and 14 were obtained.
  • a fourth isoform preparation used as its starting material erythropoietin containing isoforms 3-9 (prepared as described above). Prior to preparative isoelectric focusing carried out essentially as
  • ampholytes (Pharmalyte 2.5-5) were pre-fractionated in a Rotofor (Bio-Rad, Richmond, CA) liquid phase isoelectric
  • the prefractionation was carried out by mixing 6.7 mL of Pharmalyte 2.5-5 with 15 g of urea and adding purified water to bring the volume to 50 mL.
  • Ampholytes were removed from the isoforms using a Centrieluter (Amicon, Danvers, MA) and a
  • the isoforms were then buffer exchanged into 0.1 M sodium chloride by gel filtration using Sephadex G-25 (Pharmacia). Analytical isoelectric focusing of the five resulting pools showed them to contain isoforms 4,5,6,7, and 8. Isoform 4 ran as several bands, indicating that it may have undergone some degradation.
  • the fifth isoform preparation was modified by the addition of a pre-focusing step to the flat bed isoelectric focusing procedure. In this modification, the protein was not added to the ampholyte/urea/gel mixture prior to electrophoresis but was added to the isoelectric focusing apparatus following generation of the pH gradient in the gel bed.
  • the pre-focusing modification was undertaken to make the ultraviolet absorbance characteristics of the isoform preparations more similar to that of the starting recombinant erythropoietin. This improvement in spectral characteristics can be seen in the ratio of absorbance at 280 and 260 nm for the isolated isoforms.
  • the average ratio of absorbance at 280 nm to that at 260 nm (A 280 /A 260 ) for isoforms from preparations 2 and 3 (non-prefocused) is 1.36 ⁇ 0.11 while the average
  • a 280 /A 260 ratio for preparations 5 and 6 is 1.68 ⁇ 0.20.
  • the average A 280 /A 260 ratios are 1.39 ⁇ 0. 11 and 1 .74 ⁇ 0.09 for preparations 2 & 3 and 5 & 6, respectively.
  • Isoform 14 may have the most atypical spectrum because it is present in the smallest amounts and is thus more subject to interferences by trace contamination by ampholyte components or because it is nearest to the electrode during the flat bed isoelectric focusing procedure).
  • Example 2 of Lai et al. (modified as described earlier by using Q-Sepharose as the anion exchange resin) is 1.91 ⁇ 0.04.
  • the starting material for isoform preparation #6 was a recombinant erythropoietin preparation containing isoforms 4-13.
  • the ampholytes were pre-focused in the Rotofor apparatus as per the fourth preparation. Ampholyte fractions having measured pHs of between 3.7 and 4.8 were used for the flat bed isoelectric focusing.
  • the flat bed was pre-focused as in run #5 and isoforms 9,10,11,12 and 13 were obtained after ultrafiltration (Centricon-10) to remove carrier ampholytes.
  • sialic acid residues are cleaved from the glycoproteins by hydrolysis with 0.35 M sulfuric acid at 80°C for 30 minutes and the solutions are neutralized with sodium hydroxide prior to analysis. In order to estimate the amount of
  • Isoforms are designated according to the number of sialic acids per molecule and range from least acidic (Isoform 9) to most acidic (Isoform 13). Isoforms 9-13 are shown in gel lanes 6-10 of Figure 1. Quantities of Isoform 14 are insufficient to accurately measure the sialic acid content.
  • the sialic acid content of this isoform is inferred from its migration on IEF gels relative to other isoforms.
  • the sialic acid content of isoforms 5-8 (preparation #4) has not been measured but is likewise inferred from their migration on IEF gels.
  • the isoforms isolated as described in Example 1 are assayed by absorbance at 280 nm, by
  • Quantitation of the amount of erythropoietin protein present using a radioimmunoassay for erythropoietin produced results having higher relative in vivo specific activity for certain isoforms because of an apparent decreased immunoreactivity of isoforms containing large amounts of sialic acid leading to an underestimation of the erythropoietin concentration and thus an
  • n is the number of independent isoform preparations which contribute to the specific activity value. In most cases several in vivo assays were performed on each isoform preparation. The same in vivo data contribute to the specific activity
  • Erythropoietin purified according to the procedures described in Lai et al., supra and containing a mixture of isoforms 9 to 14 is used as a standard for the RIAs and in vivo assays.
  • the relative specific activities expressed as units/mg erythropoietin polypeptide can be converted to units/A 280 by multiplying by 0.807 mg erythropoietin polypeptide/A 280 .
  • the conversion factor is derived by multiplying the extinction coefficient of erythropoietin (1.345 mg/A 280 ) by the protein content of the
  • erythropoietin glycoprotein (about 60% by weight, Davis et al. Biochemistry 26, 2633 (1987)) to obtain mg erythropoietin polypeptide/A280 (i.e., 1.345 mg erythropoietin/A 280 ⁇ 0.60 mg polypeptide/mg
  • erythropoietin 0.807 mg polypeptide/A 280 ).
  • specific activities expressed as units/mg erythropoietin polypeptide can be multiplied by the factor 0.60 mg polypeptide/mg erythropoietin
  • glycoprotein to give specific activities expressed as units/mg erythropoietin glycoprotein.
  • Cell conditioned media from the production of recombinant erythropoietin according to the procedures described in Lin, supra are concentrated and diafiltered against 10 mM Tris, pH 7.2. Protein concentration is determined by the Bradford microprotein assay using bovine serum albumin as a standard. 19.6 ml of the solution containing 40 mg of total protein is made 20 ⁇ M in CUSO 4 , filtered through a 0.45 micron cutoff filter and loaded onto a 4 ml bed volume (1.05 cm height ⁇ 2.2 cm diameter) column packed with Q Sepharose Fast Flow (Pharmacia) which has been equilibrated with 10 mM Tris, pH 6.8 to 7.0 at 4°C. After sample application, the column is washed with two column volumes of the same buffer. The column flow rate is about 1 ml/min. Six separate columns are set up using this procedure to select defined erythropoietin isoform mixtures.
  • the eluted isoform pools from each column are concentrated and solvent exchanged into water using an Amicon Centricon-10 microconcentrator.
  • the results of analytical isoelectric focusing of these concentrated pools are shown in Figure 3.
  • Gel lanes 1-6 represent defined erythropoietin isoform mixtures eluted from column 1-6, respectively.
  • the "isoform mixture" shown in the far right gel lane of Figure 3 represents cell media which is applied to a Q-Sepharose column as described above, the column is washed with 5 mM acetic acid, 1 mM glycine, 20 ⁇ M CuSO 4 , 6M urea, and the erythropoietin isoform mixture is eluted from the column using the procedures described above.
  • sialic acids containing from approximately 8 to approximately 12 sialic acids are eluted from the column using a gradient starting at approximately 2 mM acetic acid in 6 M urea/1 mM glycine/20 ⁇ M CUSO4 and running to 40 mM acetic acid/6 M urea/1 mM glycine/20 ⁇ M CUSO 4
  • the total volume of the gradient is approximately 40 column volumes and fractions of approximately one column volume each are collected into vessels containing a volume of Tris buffer sufficient to bring the pH into the range of 6-8.5 so as to avoid long term exposure of the collected fractions to low pH.
  • Figure 4 shows the separation of isoforms 8-11 which may be achieved by this procedure.
  • Isoforms 12-14 which remain bound to the column at the end of the gradient are eluted by washing with a buffer consisting of 10 mM TrisHCl, 140 mM NaCI, 20 mM CuSO 4 (pH 7.0).
  • the isoforms (separated during the gradient or eluted by the sodium chloride solution) are freed of contaminating proteins by reverse phase chromatography followed by gel
  • oligonucleotide primers were synthesized for use in in vitro mutagenesis:
  • EPO 5' GGGCCTGGCCAACCTGTCGGAAG 3' (SEQ ID. NO: 3)
  • EPO 5' TCCCCTCCAGATAATGCCTCAGCTGC 3' (SEQ ID. NO: 4)
  • EPO 5' CAGATGCGAACTCATCTGCTCCAC 3'
  • EPO 5' AGGCCTGCAGGAATGGGAGCAGATGACCAGGTG 3' (SEQ ID. NO: 6)
  • EPO 5' TCCAGATGCGACCTCAGCTGCTC 3' (SEQ ID. NO: 7)
  • EPO was constructed to add an N-glycosylation site at Asn 69.
  • Ser 127 EPO was constructed to add an N-glycosylation site at Asn 125.
  • Thr 125 EPO and [Pro 124 , Thr 125 ] EPO were constructed to add an
  • oligonucleotide primers were synthesized for use in in vitro mutagenesis:
  • the oligonucleotide primer 5' AGATCCGACCACCGCTGCTCCAC 3' (SEQ ID. NO: 16) is used to generate [Pro 124 , Thr 125 , Thr 126 ] EPO.
  • the oligonucleotide primer 5' AGATCCGACCACCGCTGCTCCAC 3' (SEQ ID. NO: 16) is used to generate [Pro 124 , Thr 125 , Thr 126 ] EPO.
  • the oligonucleotide primer 5' AGATCCGACCACCGCTGCTCCAC 3' (SEQ ID. NO: 16) is used to generate [Pro 124 , Thr 125 , Thr 126 ] EPO.
  • the oligonucleotide primer 5' AGATCCGACCACCGCTGCTCCAC 3' (SEQ ID. NO: 16) is used to generate [Pro 124 , Thr 125 , Thr 126 ] EPO.
  • 5'TGCTCCACTCACAACAATCACTG 3' (SEQ ID. NO: 17) is then used to generate [Pro 124 , Thr 125 , Thr 126 , Thr 131 ] EPO. [Asn 69 , Thr 71 ] EPO and [Ser 68 , Asn 69 , Thr 71 ]
  • EPO are constructed to add an N-glycosylation site at Asn 69 and to enhance N-glycosylation at that site. [Asn 125 , Thr 127 ] EPO, [Asn 125 , Thr 127 , Thr 131 ] EPO,
  • [Pro 124 , Asn 125 , Ser 127 ] EPO and [Pro 124 , Asn 125 , Thr 127 ] EPO are constructed to add an N-glycosylation site at Asn 125 and to increase glycosylation at that site .
  • [Thr 125 , Thr 126 ] EPO and [Pro 124 , Thr 125 , Thr 126 , Ser 131 ] EPO are constructed to add an O-glycosylation site at Thr 125 and to increase glycosylation at that site.
  • Plasmid DNA derived from Hu13 was digested with BstEII and BglII restriction enzymes, the resulting DNA fragments were subjected to agarose gel electrophoresis, and the 810 base pair (bp)
  • Plasmid pBRgHuEPO contains the erythropoietin genomic gene as a BamHI fragment inserted into a derivative of pBR322, as described in commonly owned Lin patent, supra.
  • pBRgHuEPO was also digested with BstEII and BglII and the 6517 bp vector fragment was recovered. Ligation of the two fragments results in IGT1.
  • pDSVL (described in commonly owned Lin patent, supra, and shown in Figure 5B) was digested with BamHI and the isolated 2.8 kilobase (kb) BamHI fragment from IGT1 containing erythropoietin cDNA was ligated into it.
  • pEC-1 was digested with BamHI and BglII and the 820 bp erythropoietin cDNA fragment was isolated. It was ligated into the BamHI site of m13mp18 to give m13-EC-1. Single stranded DNA was recovered from supernatants of E. coli strain RZ1032 infected by m13-EC-1 as described by Kunkel et al. Methods in
  • reaction volume was adjusted to 10 ⁇ l with water, the mixture was heated to 65°C for annealing of the primer to the template.
  • plaques on nutrient agar were transferred to Gene Screen filters (New England Nuclear). The filters were dried under a heat lamp and then incubated for one hour in 6x SSC containing 1% SDS at 60°C .
  • the oligonucleotide primer above (8 pmoles) was end-labeled with T4 polynucleotide kinase and ⁇ 32 P-iabeled ATP and incubated with the filters overnight in 6x SSC, 0.5% SDS and 100 mg/ml salmon sperm DNA at 37°C for the [Asn 124 ] mutation, 55°C for the
  • pEC-1 was digested with BstEII followed by a partial digestion with BglII and the 5' termini of the resulting fragments are dephosphorylated with bacterial alkaline phosphatase in 10 mM Tris, pH 8 at 60°C for 60 minutes.
  • the 7 kb vector fragment lacking the 810 bp BstEII-BglII fragment was isolated and ligated to the erythropoietin fragments above.
  • the resulting plasmids (designated pEC-X where X is the analog number) contain DNA encoding erythropoietin analogs having altered amino acid residues at the indicated positions.
  • an analog of erythropoietin was constructed by in vitro mutagenesis that deleted amino acid residues 41-55. This resulted in a smaller (775 bp) EPO containing BstEII-BglII fragment. The fragment was inserted into pECl as described above.
  • pEC34 was digested with BstEII, partially digested with BglII, dephosphorylated and the vector isolated as described above. The 7kb vector fragment was then ligated to erythropoietin fragments as described above. Cloning with pEC34 allows easy discrimination between
  • Reclosures result in a smaller BstEII-BglII fragment than analogs and these can be easily differentiated on agarose gels.
  • Plasmids designated pDEC-X were constructed by inserting erythropoietin cDNA into pDEC ⁇ , which is a derivative of plasmid pDS ⁇ 2.
  • the expression vector pDS ⁇ 2 is generally described in
  • pDEC ⁇ was derived from pDS ⁇ 2 by the following steps:
  • the HindIII site of pDS ⁇ 2 was deleted by digesting pDS ⁇ 2 DNA with HindIII, treating the HindIII cohesive ends with E. coli DNA Polymerase (Klenow fragment) and dNTPs, and religating the blunt-ended vector.
  • the resulting plasmid was pDS ⁇ 2 ⁇ H.
  • pDS ⁇ 2 ⁇ H was digested with Sail and a synthetic oligonucleotide having an SV40 splice signal with a Sail linker attached to the 3' end of the splice signal was ligated to it.
  • the synthetic oligonucleotide had the following sequence (SEQ ID. NO: 18) :
  • pDS ⁇ 2 ⁇ H splice was digested with SalI and blunt-ended by treating the cohesive ends with T4 DNA polymerase and dNTPs.
  • pDEC was digested with KpnI and PvuII and blunt-ended by treating the cohesive ends with mung bean nuclease.
  • the plasmid was religated to delete the excised KpnI-PvuII fragment resulting in the plasmid pDEC ⁇ .
  • pDEC-X plasmids were made from pDEC ⁇ by complete digestion with BstEII followed by a partial digestion with BglII.
  • the vector fragment lacking erythropoietin coding sequences was isolated and ligated to the 810 bp BstEII-BglII fragments containing the desired plasmid.
  • pDEC(N47) and pDEC(N48) pDEC(N47) which contains asn30 thr32 val87 asn88 and thr90 mutations was constructed from pDEC(N18) and pDEC(N4).
  • pDEC(N18) was digested with HindII and BglII and a 445 bp fragment was isolated.
  • pDEC(N4) was digested with BstEII and HindIII and a 377 bp fragment was isolated. These two fragments were ligated into pDEC ⁇ cut with BstEII and BglII as described above resulting in pDEC(N47).
  • pDEC(N48) which contains asn69 thr71 ser87 asn88 and thr90 mutations was constructed from pDEC(N14) and pDEC(N11).
  • pDEC(N14) was digested with Hindi! and BglII and a 445 bp fragment was isolated.
  • pDEC(Nll) was digested with BstEII and HindII and a 377 bp fragment was isolated. These two fragments were ligated into pDEC ⁇ cut with BstEII and BglII as described above
  • pDEC(062) (HCG-Erythropoietin Fusion)
  • pDEC(062) was assembled from pECl and a 107 base pair StuI-BglII synthetic DNA linker containing the carboxy terminal 28 amino acids from human chorionic gonadotropin (ser-ser-ser-lys-ala-pro-pro-pro-ser-leu-pro-ser-pro-ser-arg-leu-pro-gly-pro-ser-asp-thr-pro-ile-leu-pro-gln) (SEQ ID. NO: 25) (Pierce et al. Ann.
  • pECl was digested with StuI and BglII and the 610bp DNA fragment was isolated.
  • the synthetic linker was phosphorylated with ATP and polynucleotide kinase and ligated with the pECl fragment into pDEC ⁇ previously digested with BstEII and partially digested with BglII as described above.
  • Construction of pDEC(NO1) pDEC(NO1) was assembled from pDEC(062) (HCGEPO) and pDEC(N14) (Ser 87 Asn 88 Thr 90 ).
  • pDEC177 was digested with StuI and BglII and the 610bp DNA fragment
  • pDEC(062) was digested with StuI and BglII and the 107 base pair fragment was isolated. These two DNA fragments were ligated into pDEC ⁇ previously digested with BstEII and partially digested with BglII as described above.
  • pDEC(NO2) pDEC(N02) was assembled from pDEC(062) (HCG- EPO) and pDEC(N47) (Asn 30 Thr 32 Val 87 Asn 88 Thr 90 ).
  • pDEC(N47) was digested with StuI and BglII and the 610bp DNA fragment containing the Asn 30 Thr 32 Val 87 Asn 88 Thr 90
  • pDEC(O62) was digested with StuI and BglII and the 107 base pair fragment was isolated. These two DNA fragments were ligated into pDEC ⁇ previously digested with BstEII and partially digested with BglII as described above. Construction of PDEC(N16) (Ser 87 Asn 88 Thr 90 Ala 162 ) pDEC(N16) was assembled from
  • pDEC(N14) (Ser 87 Asn 88 Thr 90 ) and pDEC258 (Ala 162 ).
  • pDEC258 was constructed using procedures for in vitro
  • pDEC(N14) was digested with StuI and BglII and the 610bp DNA fragment containing the Ser 87 Asn 88 Thr 90 mutations was isolated with GeneCleanTM.
  • pDEC258 was digested with StuI and BglII and a 210 base pair fragment was isolated. These two DNA fragments were ligated into pDEC ⁇ previously digested with BstEII and partially digested with BglII as described above.
  • pDEC(R1) (gln 24 ser 87 asn 88 thr 90 )
  • DEC(R2) (gln 38 ser 87 asn 88 thr 90 )
  • pDEC(R3) gln 83 ser 87 asn 88 thr 90
  • pEC9 (gln 83 ) was constructed using the primer
  • a volume of supernatant containing 5-20 units from COS cells transfected with erythropoietin analog cDNAs as described in Example 6 was immunoprecipitated overnight at room temperature with a rabbit antierythropoietin polyclonal antibody. 20-80 ⁇ l of 1:1 Protein A-Sepharose in phosphate buffered saline (PBS) was added to the immunoprecipitate and allowed to incubate for one hour at room temperature. The samples were centrifuged, washed with PBS and, where indicated, the pellet was treated with N-glycanase to remove
  • PBS phosphate buffered saline
  • RIAs were performed according to Egrie et al., supra. EIAs were performed with CLINIGENTM EIA kit (R and D Systems) using procedures supplied by the manufacturer. The in vivo biological activity of erythropoietin analogs was determined on supernatants from CHO cells expressing an erythropoietin analog or on purified erythropoietin obtained from CHO cell
  • the mononucleated cells from human bone marrow cells were partially purified on a ficoll-paque cushion and washed in Iscove medium before plating to remove the adherent cells.
  • the culture medium contained 0.9% methyl cellulose and did not include any bovine serum albumin.
  • the erythropoietin analogs transfected and expressed in COS cells as described in Example 6 were analyzed in crude COS cell supernatants using RIA, EIA and an erythroid colony forming assay.
  • Purified human sequence erythropoietin has an in vitro activity that was comparable to the RIA activity as determined by the above-mentioned assays.
  • HCG C-terminal extension 3 at least 3g 1.0-1.3
  • HCG extension 4 at least 3g 1.5
  • dO-linked chain at Ser 126 is present on 70% of human erythropoietin molecules.
  • eAnalogs having reduced O-glycosylation have a carbohydrate chain at Ser 126 on less than 70% of the molecules.
  • fThr 12 3 EPO has two O-linked chains on greater than 60% of the molecules.
  • Thr 125 EPO has two O-linked chains on about 40% of the molecules.
  • Pro 124 Thr 125 EPO has two O-linked chains on greater than 80% of the molecules.
  • Ratio of in vivo activity , to RIA or EIA was determined as described in footnote a of Table 6.
  • the erythropoietin analog [Thr 125 ] EPO was constructed as described in [Section A] Example 6.
  • [Thr 125 ] mutation was isolated by cleaving the plasmid pEC containing the [Thr 125 ] mutation with BstEII and BglII and ligating the fragment to pDEC ⁇ , [a derivative of pDS ⁇ 2] (described in Example 6).
  • erythropoietin cDNA was transfected into DHFR-deficient CHO cells.
  • 770 ml of CHO cell conditioned medium was concentrated using a 10,000 dalton molecular weight cutoff membrane and diafiltered against 10 mM Tris-HCl, pH 8.6 to a final volume of 34 ml.
  • a 17 ml. aliquot of the concentrate was loaded onto a Q-Sepharose fast flow column (5 ml bed volume) equilibrated in the same buffer and eluted in a linear gradient of 0-250 mM NaCI in 10 mM Tris-HCl, pH 8. 6.
  • Aliquots of column fractions, either untreated or digested with N-glycanase were analyzed by SDS-PAGE or IEF and pools (designated 2, 3 and 4) were made based upon the isoform and/or
  • the EPO N14 analog was purified using a three step procedure consisting of ion exchange
  • Conditional medium from CHO cells expressing the EPO N14 analog was harvested and
  • the concentrated medium was loaded at ⁇ 12 A 280 /ml resin to Q-Sepharose FF (Pharmacia) column equilibrated in 10mM Tris pH 8.5 at a flow rate of 0.2 cm/min.
  • the isoform pool was loaded at ⁇ 5 A 280 /ml resin to a reverse phase C4 column (Vydac) equilibrated in 20% ethanol/10mM Tris pH 7.0 at a flow rate of 2.5 cm/min.
  • the column was washed with 1CV of 20%
  • Pool #1 was concentrated approximately 65-fold and pool #2 approximately 250-fold using Centricon 10's (Amicon). Each pool was buffer exchanged into 20mM NaCitrate/100mM NaCI pH7.0.
  • EPO N14 analog was purified using a three step procedure consisting of ion exchange chromatography, reverse phase HPLC, and hydroxylapatite chromatography.
  • the concentrated medium was loaded at ⁇ 10 A 280 /ml resin to Q-Sepharose FF (Pharmacia) column equilibrated in 10mM Tris pH 8.5 at a flow rate of 0.2 cm/min.
  • IEF gel pH 3-5 Three separate fraction pools were prepared based on the isoform distribution as determined by IEF. A low isoform pool (isoform 4-12), a medium isoform pool (isoform 5-15), and a high isoform pool (isoform 6-18) were made.
  • Each sample was diluted to 2 A 280 /ml in 10mM Tris pH 7.2 and an equal volume of 4M Guanidine HCI (GuHCl), 10mM CHAPS, 10mM Tris pH 7.2 was added for a final sample concentration of 1 A 280 /ml.
  • Each sample was loaded onto a hydroxylapatite minicolumn equilibrated with 2M GuHCl, 5mM CHAPS, 10mM Tris pH 7.2. The column was washed with equilibration buffer and 1 CV fractions of flow through were collected.
  • the final pools were analyzed by SDS-PAGE and IEF gels.
  • the IEF gels are shown in Figure 12.
  • the RIA and in vivo activity assays were performed as described in Example 7A.
  • the in vivo activity of the final isoform pools is reported in Table 8.
  • the high sialic acid isoform pool was used in experiments to determine the increase in the hematocrit of mice. TABLE 8
  • EPO N14 isoform pools were prepared as described in Example 7C. rHuEPO was prepared
  • rHuEPO isoform 14 was purified as follows: rHuEPO consisting of a mixture of isoforms 10, 11, 12, 13, 14 and 15 was loaded to a
  • Sprague-Dawley rats weighing between 310-378g. At various time points after administration, 0.3ml of blood was collected and serum was prepared by centrifugation. The 125 I-EPO (either recombinant human, isolated isoform 14, or EPO N14 analog) concentration in 0.1ml of each serum sample was then determined following an overnight 4°C incubation with 90% ethanol. The ethanol-precipitated 125 I-EPO in each serum sample was counted in a gamma counter and the resulting pharmacokinetic curves are shown in figure 13. The pharmacokinetic parameters were determined for each rat using PCNONLIN 4.0
  • erythropoietin group also had the fastest clearance half-life of 1.78 hrs compared to 2.40 hrs for isolated isoform 14 and 3.03 hrs for EPO N14 analog.
  • EPO N14 analog isoforms 15-17
  • erythropoietin receptor was studied by a cold displacement assay using human erythroleukemia 0CIM1 cells (Papayannopoulou et al. Blood 64 (supp.l), 116a (1984)).
  • Increasing concentrations of unlabeled EPO N14 were incubated with OCIM1 cells along with a constant concentration of 125 I-rHuEPO to determine the amount of EPO N14 required to compete with 125 I rHuEPO for binding to the receptor.
  • OCIM1 cells Chrot al. 1 cells
  • Unlabeled EPO N14 was diluted in assay buffer and added in amounts of 0.03ng, 0. lng, 0.3ng, 1.0ng, 3.0ng, 10.0ng, and 30.0ng (based on peptide mass).
  • CD1 mice (about 30g) were injected
  • erythropoietin preparations was based on peptide mass, so that a 30g mouse received either 0.071 ⁇ g of
  • mice high pool and isoform 14. The hematocrits of all mice were determined at baseline and twice weekly thereafter by retroorbital bleeds. At the conclusion of the experiment, serum from all animals was collected and assayed for antibodies to the injected product. Data from animals judged to be negative for neutralizing antibodies was used for subsequent analysis.
  • mice treated with the EPO N14 high isoform pool attained the highest group average hematocrits compared to the other preparations.
  • Isoform 14, recombinant human EPO and the EPO N14 low isoform pool increased hematocrit to a lesser extent.
  • EPO N14 high isoform pool appears to be more active on a peptide mass basis than any other preparation tested. Both the EPO N14 high isoform pool and isoform 14 were more active than recombinant human EPO.
  • the EPO N14 low pool had the lowest activity when any of the analyses was used for comparison.
  • 3Total hematocrit increase was calculated as the group average hematocrit on day 39 minus group average hematocrit at baseline.
  • MOLECULE TYPE protein

Abstract

Erythropoietin analogs having at least one additional site for glycosylation, or a rearrangement of at least one site for glycosylation are disclosed. The invention also relates to DNA sequences encoding said erythropoietin analogs, and recombinant plasmids and host cells for analog expression.

Description

ERYTHROPOIETIN ANALOGS
This is a continuation-in-part application of U.S. Serial No. 07/942,126, filed September 8, 1992. The present invention relates to erythropoietin analogs having at least one additional site for glycosylation or a rearrangement of at least one site for glycosylation. The invention also relates to DNA sequences encoding said erythropoietin analogs, and recombinant plasmids and host cells for analog expression.
Background of the Invention
Erythropoietin (EPO) is a glycoprotein hormone involved in the maturation of erythroid progenitor cells into erythrocytes. It is essential in regulating levels of red blood cells in circulation. Naturally occurring erythropoietin is produced by the liver during fetal life and by the kidney of adults and circulates in the blood and stimulates the production of red blood cells in bone marrow. Anemia is almost invariably a
consequence of renal failure due to decreased production of erythropoietin from the kidney. Recombinant
erythropoietin produced by genetic engineering
techniques involving the expression of a protein product from a host cell transformed with the gene encoding erythropoietin has been found to be effective when used in the treatment of anemia resulting from chronic renal failure.
Low levels of erythropoietin are normally present in human urine, while individuals suffering from aplastic anemia exhibit elevated levels of urinary erythropoietin. The purification of human urinary erythropoietin by Miyake et al. in J. Biol. Chem., 252, 5558 (1977), used, as starting material, urine from aplastic anemic individuals. To date, however, urinary erythropoietin has not been shown to be therapeutically useful.
The identification, cloning, and expression of genes encoding erythropoietin are described in U. S. Patent No. 4,703,008 to Lin, the disclosure of which is incorporated herein by reference. A description of a method for purification of recombinant erythropoietin from cell medium is included in U. S. Patent No.
4,667,016 to Lai et al. The expression and recovery of biologically active recombinant erythropoietin from mammalian cell hosts containing the erythropoietin gene on recombinant plasmids has, for the first time, made available quantities of erythropoietin suitable for therapeutic applications. In addition, knowledge of the gene sequence and the availability of larger quantities of purified protein has led to a better understanding of the mode of action of this protein.
Many cell surface and secretory proteins produced by eucaryotic cells are modified with one or more oligosaccharide groups. This modification,
referred to as glycosylation, can dramatically affect the physical properties of proteins and can also be important in protein stability, secretion, and
subcellular localization. Proper glycosylation can be essential for biological activity. In fact, some genes from eucaryotic organisms, when expressed in bacteria (e.g., E. coli) which lack cellular processes for glycosylating proteins, yield proteins that are
recovered with little or no activity by virtue of their lack of glycosylation.
Glycosylation occurs at specific locations along the polypeptide backbone and is usually of two types: O-linked oligosaccharides are attached to serine or threonine residues while N-linked oligosaccharides are attached to asparagine residues when they are part of the sequence Asn-X-Ser/Thr, where X can be any amino acid except proline. The structures of N-linked and O-linked oligosaccharides and the sugar residues found in each type are different. One type of sugar that is commonly found on both is N-acetylneuraminic acid
(hereafter referred to as sialic acid). Sialic acid is usually the terminal residue of both N-linked and
O-linked oligosaccharides and, by virtue of its negative charge, may confer acidic properties to the
glycoprotein.
Both human urinary derived erythropoietin and recombinant erythropoietin (expressed in mammalian cells) having the amino acid sequence 1-165 of human erythropoietin contain three N-linked and one O-linked oligosaccharide chains which together comprise about 40% of the total molecular weight of the glycoprotein.
N-linked glycosylation occurs at asparagine residues located at positions 24, 38 and 83 while O-linked glycosylation occurs at a serine residue located at position 126 (Lai et al. J. Biol. Chem. 261, 3116
(1986); Broudy et al. Arch. Biochem. Biophys. 265, 329 (1988)). The oligosaccharide chains have been shown to be modified with terminal sialic acid residues.
Enzymatic treatment of glycosylated erythropoietin to remove all sialic acid residues results in a loss of in vivo activity but does not affect in vitro activity
(Lowy et al. Nature 185, 102 (1960); Goldwasser et al. J. Biol. Chem. 249, 4202 (1974)). This behavior has been explained by rapid clearance of asialoerythropoietin from circulation upon interaction with the hepatic asialoglycoprotein binding protein (Morrell et al. J. Biol. Chem. 243. 155 (1968); Briggs, et al. Am. J. Physiol. 227. 1385 (1974); Ashwell et al. Methods Enzymol. 50, 287 (1978)). Thus, erythropoietin
possesses in vivo biological activity only when it is sialylated to avoid its binding by the hepatic binding protein. The role of the other components in the oligosaccharide chains of erythropoietin is not well defined. It has been shown that partially
deglycosylated erythropoietin has greatly reduced
in vivo activity compared to the glycosylated form but does retain in vitro activity (Dordal et al .
Endocrinology 116, 2293 (1985) ; Lin patent, supra) . In another study, however, the removal of N-linked or O-linked oligosaccharide chains singly or together by mutagenesis of asparagine or serine residues that are glycosylation sites sharply reduces in vitro activity of the altered erythropoietin that is produced in mammalian cells (Dube et al . J. Biol . Chem. 263, 17516 (1988) ) .
Glycoproteins such as erythropoietin can be separated into different charged forms using techniques such as isoelectric focusing (IEF) . Several parties have reported IEF studies of crude and partially purified erythropoietin preparations (Lukowsky et al . , J. Biochem 50 , 909 (1972) ; Shelton et al . Biochem. Med . 12, 45 (1975) ; Fuhr et al . Biochem. Biophys . Res . Comm. 98, 930 (1981) ) . At most, three or four fractions having erythropoietin activity were distinguished by IEF in these studies and none were characterized with respect to carbohydrate content . In addition, no correlation between the isoelectric points of the fractions and their biological activity was made .
During the purification of urinary erythropoietin from human urine discussed in Miyake et . al . supra, two erythropoietin fractions from
hydroxylapatite chromatography designated II and IIIA were reported to have similar specific activity . A subsequent carbohydrate analysis of fractions II and IIIA revealed that fraction II had a greater average sialic acid content than fraction IIIA (Dordal et . al . supra) . It is an object of the present invention to provide separated and isolated isoforms of
erythropoietin having a defined sialic acid content and biological activity. Pharmaceutical compositions containing such molecules would have therapeutic benefit.
Summary of the Invention The subject invention relates to analogs of human erythropoietin comprising an amino acid sequence which includes at least one additional site for
glycosylation. The added sites for glycosylation may result in a greater number of carbohydrate chains, and higher sialic acid content, than human erythropoietin. Erythropoietin analogs comprising amino acid sequences which include the rearrangement of at least one site for glycosylation are also provided. Analogs comprising an addition of one or more amino acids to the carboxy terminal end of erythropoietin wherein the addition provides at least one glycosylation site are also included. The invention further encompasses DNA
sequences encoding such erythropoietin analogs, and recombinant plasmids and host cells for analog
expression.
Brief Description of the Drawings
Figure 1 shows an analytical isoelectric focusing gel of the separate recombinant erythropoietin isoforms. Gel lanes 1-11 show isoforms ranging from less acidic (higher pi) in lane 1 to more acidic (lower pI), in lane 11. Purified recombinant erythropoietin containing a mixture of isoforms 9-14 is also shown in the far left and right lanes of the gel. Figure 2 shows the relationship between the number of sialic acids per erythropoietin isoform and the in vivo specific activity of each isoform expressed as units per mg of erythropoietin polypeptide. In
Figure 2A, the concentration of each erythropoietin isoform was determined by the Bradford protein assay; in 2B, the concentration was determined by absorbance at 280 nm, in 2C, the concentration was determined by RIA.
Figure 3 shows an analytical isoelectric focusing gel of defined mixtures of recombinant
erythropoietin isoforms prepared by anion exchange chromatography under different conditions. Gel lanes 1-6 represent, respectively, erythropoietin isoforms eluted in a high salt wash after washing the Q-Sepharose fast flow column with 150 mM acetic acid, pH 4.7, 150 mM acetic acid (unbuffered), 200 mM acetic acid, pH 4.7, 250 mM acetic acid, pH 4.7, 300 mM acetic acid, pH 4.7 or 300 mM acetic acid (unbuffered). Purified
recombinant erythropoietin containing a mixture of isoforms as obtained using procedures described in
Example 2 of Lai et al., supra, except that DEAE-Agarose chromatography is replaced by Q-Sepharose
chromatography, is also shown in the far left lane of the gel.
Figure 4 shows the separation of
erythropoietin isoforms 8 to 12 achieved by subjecting cell conditioned medium applied to a column of
Q-Sepharose to a gradient of decreasing pH and
increasing ionic strength. Aliquots of even numbered fractions from Fraction 2 to Fraction 40 were subjected to analytical isoelectric focusing. Purified
recombinant erythropoietin containing a mixture of isoforms obtained using procedures described in
Example 2 of Lai et al. supra, except that DEAE-Agarose chromatography is replaced by Q-Sepharose chromatography, is also shown in the far left lane of the gel.
Figure 5 shows the amino acid sequence of human erythropoietin. Squares indicate asparagine residues to which N-linked carbohydrate chains are attached and an asterisk indicates the serine residue modified with an O-linked carbohydrate chain.
Figures 6A, 6B, and 6C show the series of cloning steps used in generating plasmids for the construction and analysis of analogs of human
erythropoietin. These analogs have amino acids altered as shown in Figure 5 which provide additional
glycosylation sites.
Figure 7 shows a Western blot analysis of COS cell supernatants of human sequence erythropoietin and indicated erythropoietin analogs. The analogs [Asn9, Serli] EPO, [Asn69] EPO, [Asn125, Ser127] EPO, and
[Pro124, Thr125] EPO are constructed as described in Example 6. Additional analogs [Pro125, Thr127] EPO and [Asni26, Seri28] EPO which do not contain additional carbohydrate chains are shown for comparison.
Figure 8 shows a Western blot analysis of COS cell supernatants of human sequence erythropoietin and indicated erythropoietin analogs after treatment with N-glycanase. The analogs [Thr125] EPO and [Pro124,
Thr125] EPO were constructed as described in Example 6. The analogs [Val126] EPO, [Pro124] EPO, [Pro125] EPO, [Thr127] EPO, [Pro125, Ser127] EPO and [Thr125, Ser127] EPO are shown for comparison.
Figure 9 shows an isoelectric focusing gel of pools 2, 3 and 4 obtained by Q-Sepharose and C4 reverse phase chromatography of cell medium that supported the growth of CHO cells transfected with erythropoietin cDNA containing the [Thr125] mutation. Purified recombinant erythropoietin containing a mixture of isoforms were obtained using procedures described in Example 2 of Lai et al., supra, except that DEAE-Agarose chromatography is replaced by Q-Sepharose chromatography, is also shown in the left and right lanes of the gel.
Figure 10 shows a Western blot analysis of COS cell supernatants of recombinant human erythropoietin (rHuEPO) and selected analogs. The construction of the analogs is described in Example 6. Analogs N9, N14, N18, N19, N21, N24 and N39 have at least one additional carbohydrate chain as evidenced by slower gel mobility.
Figure 11 shows a Western blot analysis of COS cell supernatants of recombinant human erythropoietin and EPO N14 analog during N-glycanase digestion. Time points were taken at 0, 4, 12 and 45 minutes and after overnight digestion.
Figure 12 shows an isoelectric focusing gel of
EPO N14 analog isoform preparations. The low isoform pool contains EPO N14 analog having mostly 6-12 sialic acids per molecule, the medium isoform pool contains analog N14 having mostly 10-15 sialic acids per
molecule, and the high isoform pool contains EPO N14 analog having mostly 12-17 sialic acids per molecule.
Figure 13 shows the pharmacokinetics of recombinant human erythropoietin, isolated isoform 14 and EPO N14 analog (isoforms 15-17) after intravenous injection into rats.
Figure 14 shows a cold displacement assay of 125I labeled recombinant human erythropoietin binding to the erythropoietin receptor in the presence of varying amounts of unlabeled rHuEPO, isolated isoform 14 or EPO N14 analog.
Figure 15 shows a mouse hematocrit study comparing the activity of EPO N14 high isoform pool (0.036 and 0.0712 μg), isolated isoform 14 (0.036 and 0.0712 μg), EPO 177 low isoform pool (0.0712 μg) and rHuEPO (0.071 μg). Detailed Description of the Invention
The subject invention provides erythropoietin isoforms. The specific isoforms of erythropoietin obtained in accordance with the present invention, and their properties, may vary depending upon the source of the starting material. For example, the isoforms of urinary derived human erythropoietin are different than the isoforms of recombinant erythropoietin. In a preferred embodiment, the invention relates to an erythropoietin isoform having a specific number (i.e. a fixed number greater than 0) of sialic acids per erythropoietin molecule, said number selected from the group consisting of 1-14. Advantageously said number is 9, 10, 11, 12, 13, or 14. In another embodiment, said number is greater than 14, advantageously 16-23.
The term "erythropoietin isoform" as used herein refers to erythropoietin preparations having a single isoelectric point (pI), and having the same amino acid sequence. The term "erythropoietin", as used herein, includes naturally occurring erythropoietin, urinary derived human erythropoietin as well as nonnaturally occurring polypeptides having an amino acid sequence and glycosylation sufficiently duplicative of that of naturally occurring erythropoietin to allow possession of in vivo biological properties of causing bone marrow cells to increase production of
reticulocytes and red blood cells.
It has been found that discrete isoforms of recombinant erythropoietin having the amino acid
sequence of urinary derived human erythropoietin
correspond to erythropoietin molecules having from 1-14 sialic acids, and each isoform present in purified recombinant erythropoietin has an in vivo activity which is related to the number of sialic acids the isoform possesses. In a preferred embodiment, erythropoietin is the product of the expression of an exogenous DNA sequence that has been transfected into a non-human eucaryotic host cell, that is, in a preferred embodiment the erythropoietin is "recombinant erythropoietin".
Recombinant erythropoietin is advantageously produced according to the procedures described in commonly owned Lin U.S. Patent 4,703,008 hereby incorporated by
reference. Recombinant erythropoietin can be purified according to the general procedures described in
Example 2 of commonly owned Lai et al. U. S. Patent 4,667,016 hereby incorporated by reference, or
alternatively according to the procedure described in Example 2 of Lai et al. wherein DEAE-Agarose
chromatography is replaced by Q-Sepharose
chromatography.
Erythropoietin purified according to Example 2 of Lai et al. supra contains predominantly six isoforms when analyzed by IEF. In addition, at least one
additional isoform of greater acidity has been detected using the chromatographic procedures described in
Example 4. (This more acidic form, migrating at >14 sialic acids on an IEF gel may contain nonsialic acid negative charges as shown by the resistance of some of the charge to sialidase digestion). These isoforms differ from each other by sialic acid content. As shown in the Examples, this is demonstrated by isolating 10 of these isoforms by preparative IEF and determining the sialic acid content of five of them. Of the isoforms assayed for sialic acid content, it is found that the five isoforms contained either 9, 10, 11, 12 or 13 sialic acid residues.
There is a relationship between the relative in vivo specific activity of erythropoietin and number of sialic acid residues per erythropoietin molecule from the isoforms 5 through 11 (each isoform is designated herein by the number of sialic acids per erythropoietin molecule). Isoforms 11 through 14 have approximately the same relative in vivo specific activity. Isoforms 5-14 were assayed for in vivo activity by the exhypoxic polycythemic mouse bioassay and the amount of each isoform present is determined by Bradford protein assay, absorbance at 280 nm or by radioimmunoassay (RIA) for erythropoietin. RIA determinations (Egrie et al.
Immunobiology 172, 213, (1986)), expressed as units/ml, are divided by 212,770 units/mg erythropoietin
polypeptide, the average specific activity of purified erythropoietin as determined by RIA, to give protein concentrations of isolated isoforms or isoform mixtures expressed as mg erythropoietin polypeptide/ml. As shown in the Examples, the relative in vivo specific
activities increase step-wise from isoform 5 to isoform 11 (see Table 2).
The in vivo specific activities referred to herein are measurements of relative in vivo specific activities and are not measurements of absolute in vivo specific activities. For the purposes of this
application, the specific activities are used only to compare relative activities of isoforms that have been assayed using the same assay, using the same conditions including the same internal standard, the same type of animals, having the same analysis of the data used to calculate specific activity, the same assay for
determining protein content. It is not intended that any in vivo specific activity value reported for any isoform represents an inherent or absolute value for that isoform.
This invention also provides compositions comprising two or more erythropoietin isoforms. In one embodiment the compositions comprise a mixture of isoforms having greater than a predetermined number of sialic acids per erythropoietin molecule, e.g. greater than 11 sialic acids per erythropoietin molecule, or greater than 12 sialic acids per molecule, e . g . a mixture of isoforms 12, 13 and 14. In another
embodiment the compositions comprise mixtures of isoforms having a predetermined number of sialic acids per erythropoietin molecule, e .g. less than 12, but greater than 8 sialic acids per molecule as in, for example, a mixture of isoforms 9, 10, and 11 . The invention also provides for compositions of
erythropoietin isoforms wherein the relative amounts of the isoforms are the same or different . For example, a mixture of isoforms 9, 10 and 11 could have the isoforms present in a variety of ratios such as 1 : 1 : 1 , 2 : 3 : 1 or 20 : 20 : 1.
Advantageously, the compositions comprise mixtures of less than four isoforms, for example a mixture of isoforms 11, 12, and 13, or a mixture of 12 and 14, or a mixture of 7 and 13.
In order to produce mixtures of erythropoietin isoforms, this invention also provides methods of isolating selected erythropoietin isoforms
simultaneously. These methods include isolation of individual isoforms by techniques such as preparative isoelectric focusing or preparation of mixtures of isoforms having a predetermined number of sialic acids per molecule (for example, greater than 11 ) by
techniques such as ion exchange chromatography or chromatofocusing . All of these techniques have as their basis the separation of proteins according to charge .
In general , ion exchange chromatography and chromatofocusing involve application of either crude human erythropoietin (cell conditioned media) or
purified material to a column resin under conditions that permit binding of some or all of the erythropoietin isoforms to the resin . For crude erythropoietin
preparations, it is preferable to apply the protein to the column at about pH 7 while for purified preparations the protein can be applied to the column at pH 7 down to about pH 4 . After washing the column with buffer at about pH 4 , those erythropoietin isoforms that remain bound on the ion exchange column are eluted by
increasing the pH and the salt concentration of the buffer or by applying a gradient of decreasing pH and increasing ionic strength at about pH 4 . For
chromatofocusing, the isoforms are eluted from the column by a gradient of decreasing pH or by washing the column with a high concentration of salt .
In a preferred embodiment, individual isoforms are isolated using ion exchange chromatography . As an example, isoform 14 was isolated using ion exchange chromatography as described in Example 8.
Also encompassed by the invention are certain analogs of human erythropoietin . As used herein the phrase "analog of human erythropoietin" refers to erythropoietin with one or more changes in the amino acid sequence of human erythropoietin which result in an increase in the number of sites for sialic acid
attachment . Analogs are generated by site-directed mutagenesis having additions, deletions, or
substitutions of amino acid residues that increase or alter sites that are available for glycosylation . Such analogs may have a greater number of carbohydrate chains than human erythropoietin .
The erythropoietin analogs of the present invention comprise an amino acid sequence which includes at least one additional site for glycosylation . Analogs having levels of sialic acid greater than those found in human erythropoietin are generated by adding
glycosylation sites which do not perturb the secondary or tertiary conformation required for biological
activity . Advantageously, the analog of human
erythropoietin has 1, 2 or 3 additional sites for N-glycosylation or O-glycosylation, resulting in the addition of 1, 2, or 3 additional N-linked or O-linked carbohydrate chains. For example, a leucine at position 69 is replaced by an asparagine to give the sequence Asn-Leu-Ser, which serves as a fourth site for
N-glycosylation. Such a change can commonly provide up to four additional sialic acids per molecule. Examples of changes that generate additional O-glycosylation sites are alanine at position 125 to threonine and alanines at positions 124 and 125 to proline and threonine, respectively. Analogs may be constructed which have one or more additional N-linked and O-linked chains, for example, analogs NO1 and N02 described in Table 5. As will be appreciated by those skilled in the art, the subject invention includes many other analogs of human erythropoietin having additional sites for glycosylation.
Analogs having increased levels of carbohydrate attachment at a glycosylation site are also encompassed by the invention. Such analogs usually involve the substitution of one or more amino acids which are in close proximity to an N-linked or O-linked site. As a result of these amino acid changes, a greater proportion of erythropoietin polypeptides will have a carbohydrate modification. The glycosylation sites may be naturally occurring or generated by
mutation. For example, analog N13 does not generate an additional carbohydrate chain even though a
glycosylation site was introduced at position 88.
However, analogs N14 and N18, which have serine and valine residues, respectfully, substituted at position 87, have an additional carbohydrate chain at position 88.
Also provided by the invention are analogs which have one or more amino acids extending from the carboxy terminal end of erythropoietin and wherein the carboxy terminal extension provides at least one
additional carbohydrate site. In one embodiment, an analog was constructed by fusing the carboxy-terminal 28 amino acids of human chorionic gonadotropin (HCG) to the arginine residue at position 166 of human
erythropoietin. The HCG carboxy-terminal fragment has four sites for O-glycosylation (Kessler et al., J. Biol. Chem. 254, 7907 (1979)).
Tables 3, 4 and 5 list erythropoietin analogs which have additional sites for N-linked and/or O-linked carbohydrate chains. The analogs have the sequence Asn-X-Ser/Thr substituted at various positions in the human erythropoietin polypeptide chain to create
N-linked sites or have serine or threonine residues introduced to create O-linked sites.
Table 6 lists those analogs which add at least one additional N-linked or one additional O-linked carbohydrate chain, or add additional N-linked and
O-linked chains simultaneously, as evidenced by the migration of the glycoproteins on SDS gels (Example 7). As can be appreciated from Tables 3-6, analogs having one or more additional sites for carbohydrate attachment do not necessarily result in erythropoietin molecules having additional carbohydrate chains. For example, substitution of threonine residues at positions 123 and 125 resulted in the addition of an O-linked carbohydrate chain while substitution of serine or threonine at other positions did not result in analogs with additional O-linked chains (see Table 4). However, substitution of asparagine residues at positions 30, 51, 57, 69, 88, 89, 136 and 138 in the human erythropoietin amino acid sequence resulted in the addition of an N-linked chain at those sites. The fusion of an HCG polypeptide fragment to the arginine residue at position 166 of human erythropoietin resulted in an erythropoietin-HCG fusion molecule having at least two additional O-linked carbohydrate chains.
The erythropoietin analogs of the present invention also encompass erythropoietin having an amino acid sequence which includes a rearrangement of at least one site for glycosylation. A rearrangement of a glycosylation site as used herein refers to the deletion of one or more glycosylation sites in human
erythropoietin and addition of one or more non-naturally occurring glycosylation sites. Analogs R1, R2 and R3 are examples of such rearrangements and were constructed by deletion of the N-linked sites at positions 24, 38 or 83, respectively, and addition of an N-linked site at position 88. However, numerous other types of
carbohydrate site rearrangements are possible and the resulting analogs may or may not have a greater number of glycosylation sites compared to human erythropoietin.
Analogs R1, R2 and R3 were analyzed for in vivo biological activity and the results shown in
Table 7. The introduction of an N-linked chain at
Asn 88 restored biological activity to erythropoietin having any one of the three naturally occurring N-linked sites deleted. These results indicate that the
positions of carbohydrate chains in erythropoietin may be changed in order to generate useful analogs without significantly affecting the biological activity.
Also encompassed by the present invention are DNA sequences encoding erythropoietin analogs having additional sites for N-linked and/or O-linked chains, analogs having a rearrangement of at least one
attachment site for a carbohydrate chain, and analogs having one or more amino acids extending from the carboxy-terminal end of erythropoietin. Procedures used to introduce changes into the human erythropoietin DNA sequence for the purpose of creating and altering attachment sites for carbohydrates are disclosed in Example 6.
These erythropoietin analogs can be the product of expression of an exogenous DNA sequence, i.e., produced through recombinant DNA technology, or can be synthesized products. An exogenous DNA sequence comprises cDNA, genomic DNA or chemically synthesized DNA encoding an erythropoietin analog. Recombinant DNA plasmids and eucaryotic host cells useful for the expression of said analogs are also provided.
Expression vectors include any vector which is capable of expressing cloned DNA sequences in a eucaryotic host cell, particularly those vectors used for expression in COS and CHO cells. Examples of such vectors include plasmids pEC and pDECΔ described in Example 6 of the specification. The cultivation of COS and CHO host cells expressing erythropoietin analogs was carried out using procedures known to one skilled in the art.
Isolated isoforms and mixtures, of isoforms derived from erythropoietin analogs are obtained using the methods described above for preparing isoforms of human erythropoietin. These methods may include isoelectric focusing, ion exchange chromatography and chromatofocusing. Preferably, ion exchange
chromatography is used to prepare individual isoforms . and mixtures of isoforms derived from erythropoietin analogs.
Increasing the number of carbohydrate chains on erythropoietin, and therefore the number of sialic acids per erythropoietin molecule, may confer
advantageous properties such as increased solubility, greater resistance to proteolysis, reduced
immunogenicity, increased serum half-life, and increased biological activity.
Conditioned media from CHO cells expressing erythropoietin analogs were analyzed for in vivo biological activity and the results shown in Table 6. Several analogs tested had activity that was 3-fold or more higher than human erythropoietin. In particular, analogs having an additional N-linked carbohydrate chain at either position 30 or 88 show 2- to 3-fold higher activity than human erythropoietin while analogs having additional O-linked chains as a result of fusion of human erythropoietin to the HCG polypeptide fragment have at least 2-fold higher activity.
Two erythropoietin analogs having additional carbohydrate chains were purified and isoform mixtures having different sialic acid contents were isolated (Example 8). Thr125 and Ser87Asn88Thr90 (EPO N14) analogs were separated into three separate isoform fractions and the in vivo biological activities for each fractions was determined. The results presented in Table 8
demonstrate that EPO N14 isoform fractions having higher sialic acid content have a greater in vivo activity.
A pool of high sialic acid isoforms of EPO N14 analog and recombinant human erythropoietin (isoforms 9-14) were studied in receptor binding assays,
pharmacokinetic experiments, and experiments to
determine increase in hematocrit of mice. Results of these assays indicate that there is a direct
relationship between sialic acid content, clearance half-life, and ability to increase the hematocrit of treated mice. Thus, as shown in figures 13, 14 and 15, EPO N14 high sialic acid isoform pool had a
significantly longer in vivo half-life and promoted a greater increase in hematocrit than did either isolated isoform 14 or recombinant human erythropoietin, even though the N14 high sialic acid isoform pool did not bind as strongly to the receptor. Another embodiment of the invention relates to mammalian (e.g., Chinese Hamster Ovary, CHO) host cells which preferentially synthesize isoforms of human erythropoietin or erythropoietin analogs having greater than a specific number of sialic acids per molecule, e.g. greater than 10 sialic acids per molecule.
Erythropoietin molecules have N-linked and O-linked oligosaccharides structures which can limit the sialic acid content of the molecule. For example,
tetraantennary (four-branched) N-linked oligosaccharides most commonly provide four possible sites for sialic acid attachment while bi- and triantennary
oligosaccharide chains, which can substitute for the tetraantennary form at asparagine-linked sites, commonly have at most only two or three sialic acids attached. O-linked oligosaccharides commonly provide two sites for sialic acid attachment. Thus, erythropoietin molecules can accommodate a total of 14 sialic acid residues provided all three N-linked oligosaccharides are
tetraantennary. Mammalian cell cultures are screened for those cells that preferentially add tetraantennary chains to recombinant erythropoietin, thereby maximizing the number of sites for sialic acid attachment.
The N-linked oligosaccharides of urinary erythropoietin contain sialic acid in both an α 2,3 and an α 2,6 linkage to galactose (Takeuchi et al. J. Biol. Chem. 263, 3657(1988)). Typically the sialic acid in the α 2,3 linkage is added to galactose on the mannose α 1, 6 branch and the sialic acid in the α 2,6 linkage i's added to the galactose on the mannose α 1,3 branch. The enzymes that add these sialic acids (β-galactoside α 2,3 sialyltransferase and β-galactoside α 2, 6
sialyltransferase) are most efficient at adding sialic acid to the mannose α 1,6 and mannose α 1,3 branches respectively.
Dihydrofolate reductase (DHFR) deficient
Chinese Hamster Ovary (CHO) cells are a commonly used host cell for the production of recombinant glycoproteins including recombinant erythropoietin.
These cells do not express the enzyme β-galactoside α 2,6 sialyltransferase and therefore do not add sialic acid in the α 2,6 linkage to N-linked oligosaccharides of glycoproteins produced in these cells. (Mutsaers et al. Eur. J. Biochem. 156, 651 (1986); Takeuchi et al. J. Chromatogr. 400, 207 (1987)). Consequently,
recombinant erythropoietin produced in CHO cells lacks sialic acid in the 2,6 linkage to galactose (Sasaki et al. (1987), supra; Takeuchi et al. (1987), supra).
In another embodiment of the subject invention, human erythropoietin or an erythropoietin analog is produced in CHO cells that are transfected with a functional β-galactoside α 2, 6 sialyltransferase gene to give incorporation of sialic acid in α 2,6 linkage to galactose. The resulting isoforms will contain sialic acid having both α 2,3 and α 2,6 linkages to galactose. See Lee et al. J. Biol. Chem. 264, 13848 (1989), hereby incorporated by reference, for a
disclosure of techniques for creating modified CHO cells or other mammalian host cells.
Also comprehended by the invention are pharmaceutical compositions comprising a therapeutically effective amount of a specific isoform or mixture of isoforms together with a suitable diluent, adjuvant and/or carrier useful in erythropoietin therapy.
Pharmaceutical compositions comprising a therapeutically effective amount of an erythropoietin analog together with a suitable diluent, adjuvant and/or carrier are also encompassed. A "therapeutically effective amount" as used herein refers to that amount which provides therapeutic effect for a given condition and
administration regimen. The administration of isoforms of human erythropoietin or erythropoietin analogs is preferably by parenteral routes. The specific route chosen will depend upon the condition being treated. The administration of isoforms of human erythropoietin or erythropoietin analogs is preferably done as part of a formulation containing a suitable carrier, such as human serum albumin, a suitable diluent, such as a buffered saline solution, and/or a suitable adjuvant. The required dosage will be in amounts sufficient to raise the hematocrit of patients and will vary depending upon the severity of the condition being treated, the method of administration used and the like.
The following examples are offered to more fully illustrate the invention, but are not to be construed as limiting the scope thereof. The
erythropoietin standard used in the in vivo bioassays employed in the Examples is a recombinant erythropoietin standard that was standardized against a partially purified urinary erythropoietin standard. Thus, only relative in vivo specific activities are being measured. Also the in vivo specific activities are expressed in "units/ml", "units/mg" and units/A280" and not as
"IU/ml", "IU/mg" and IU/A280" because the erythropoietin standard employed has not been directly correlated to any existing international standard.
Example 1; Isolation of Recombinant Erythropoietin Isoforms Recombinant erythropoietin is produced as described in Lin, supra. Recombinant erythropoietin used as starting material for the first and third isoform isolations is purified according to the
procedure described in Example 2 of commonly owned Lai et al., supra. Starting material for the second and fifth isoform isolation is purified according to Lai et al. supra using the modification of Q-Sepharose
chromatography. These preparations contain a mixture of isoforms of recombinant erythropoietin having the same amino acid sequence as urinary derived human
erythropoietin and contain predominantly isoforms 9 to 14. Starting material for the fourth isoform
preparation is the material which elutes during the 5 mM acetic acid/1 mM glycine/6M urea wash of the anion exchange column in Example 2 of Lai et al. This fraction contains isoforms with less than or equal to 9 sialic acids and was further purified by gel filtration chromatography as described in Example 2 of Lai et al. prior to use in the preparative isoelectric focusing procedure. The sixth isoform preparation used as its starting material a purified preparation of recombinant erythropoietin having from 4 to 13 sialic residues.
This material was purified as per Example 2 of Lai et al. except for a modification to the ion exchange column (elution of the recombinant erythropoietin with a sodium chloride gradient at pH 8.4 and omission of the acetic acid/urea wash) which results in retention of most of the isoforms present in the starting material.
Six different preparations of individual isoforms are carried out by preparative isoelectric focusing in a granulated gel bed (Ultrodex, LKB)
essentially as per LKB Application Note 198. Pharmalyte (Pharmacia) 2.5-5 ampholytes (Pharmacia) are used and the gel bed contains 5 M urea.
In the first preparation, approximately 20 mg of recombinant erythropoietin in 6.8 ml of 20 mM sodium citrate/ 100 mM sodium chloride, pH 7.0 are applied to the gel and focused at 8 watts for approximately 16 hours. After isoelectric focusing, the isoform bands in the gel are visualized by a paper contact print of the gel bed. The print is made and then fixed by soaking in 3 changes (approximately 10 minutes each, room temperature) of fixing solution (40% methanol/10% acetic acid/10% TCA/3.5% sulfosalicylic acid), subjected to one change (approximately 10 minutes) of 40% methanol/10% acetic acid (30-60°C), stained for 15 minutes at 60°C in 0.125% Coomassie Blue R-250/40% methanol/10% acetic acid, and then destained in 7.5% methanol/10% acetic acid in order to visualize the separated isoforms. The region of the granulated gel bed containing the isoforms (~50% of the resin) is removed, water is added (~16 ml), and the slurry is poured into a 5.5 × 24.5 inch tray and evaporated to -40 g net weight. This preparation is focused for a second time and a contact print of the gel bed is made as before. The portion of gel containing each of the six discernible isoforms is removed from the gel bed.
In order to elute the isoforms from the gel, a solution containing 10 mM Tris-HCl, pH 7.0/ 5 mM Chaps is added to each isoform to generate a slurry. The slurries are placed in small columns and washed with the Tris-Chaps buffer. The flow throughs are collected and applied separately to small columns (open column
configuration) containing Vydac C4 reversed phase resin equilibrated in 20% ethanol/10 mM Tris-HCl, pH 7.0. The columns are developed stepwise with 20% ethanol/10 mM Tris-HCl, pH 7.0, 35% ethanol/10 mM Tris-HCl, pH 7.0, and 65% ethanol/10 mM Tris-HCl, pH 7.0. The fraction eluting at 65% ethanol/10 mM Tris is diluted 1:1 with 10 mM Tris-HCl, pH 7.0 and subjected to concentration and then buffer exchanged to 10 mM Tris-HCl, pH 7.0 using a Centricon-10 (Amicon) microconcentrator. Analytical isoelectric focusing of this preparation is performed essentially as described in LKB technical note 250 using Servalyte 3-5 ampholines (Serva) in a polyacrylamide gel containing 5 M urea.
In a second preparation, approximately 26 mg of recombinant erythropoietin in 6.5 ml of deionized water are applied to the gel and focused at 2.5 watts for 35 minutes and 10 watts for approximately 17 hours. The bands of focused protein, which are visible in the gel bed, are removed as 11 different pools. Each pool is brought to about 7.5 ml with deionized water and 20 ml of each of the resulting pool supernatants is
subjected to analytical isoelectric focusing as
described above. To each of the pools is added 5 ml of 1.5 M Tris-HCl, pH 8.8 and the slurries are each placed in small columns and the liquid phase allowed to flow through. The resin is washed with approximately three volumes of 0.5 M Tris-HCl, pH 7 and the rinse solution is combined with the flow through. The eluants are concentrated and buffer exchanged to 20 mM sodium citrate/ 100 mM sodium chloride, pH 7.0 using Amicon disposable ultrafiltration devices having a 10,000 dalton molecular weight cutoff. The concentrated solutions (approximately 0.5 ml) are then passed through a 0.22 micron cutoff cellulose acetate filter. Based upon analytical isoelectric focusing, five pools are found to contain predominantly the single isoforms 10, 11, 12, 13 and 14.
In a third preparation, approximately 30 mg of recombinant erythropoietin in 21.8 ml of distilled water is applied to the gel and focused at 2 watts for 25 minutes, 10 watts for 20 hours and 15 watts for 15 minutes. Protein bands corresponding to the individual isoforms are observed visually and removed from the gel bed. Distilled water is added to gel-isolated isoforms to generate a slurry and the resulting supernatants are analyzed by analytical isoelectric focusing. An equal volume of 1 M Tris-HCl, pH 7.2 is added to each slurry, the suspensions are placed into separate small columns, and the liquid phase is allowed to flow through the column to elute the isoforms. Each flow through is concentrated and buffer exchanged to 20 mM sodium citrate/ 100 mM sodium chloride, pH 7.0 using Amicon disposable ultrafiltration devices having a 10,000 dalton molecular weight cutoff. An analytical
isoelectric focusing gel revealed that pools containing predominantly the single isoforms 9, 10, 11, 12, 13 and 14 were obtained.
A fourth isoform preparation used as its starting material erythropoietin containing isoforms 3-9 (prepared as described above). Prior to preparative isoelectric focusing carried out essentially as
described for preparations 1-3 above, the ampholytes (Pharmalyte 2.5-5) were pre-fractionated in a Rotofor (Bio-Rad, Richmond, CA) liquid phase isoelectric
focusing cell to yield an ampholyte range more suitable for the lower isoelectric points of the starting
material. The prefractionation was carried out by mixing 6.7 mL of Pharmalyte 2.5-5 with 15 g of urea and adding purified water to bring the volume to 50 mL.
This mixture was fractionated in the Rotofor at
10 Watts, 1°C, for 5 1/2 hours using 0.1 M phosphoric acid and 0.1 M sodium hydroxide as the anolyte and catholyte, respectively. The ampholyte fractions having measured pHs of between 4.5 and approximately 6 were used in the flat-bed isoelectric focusing.
Ampholytes were removed from the isoforms using a Centrieluter (Amicon, Danvers, MA) and a
10,000 MW cutoff Centricon (Amicon) using the following parameters: 0.18 Tris buffer pH 8.8, 100 Volts,
25-30 mA, for 3 hours. The isoforms were then buffer exchanged into 0.1 M sodium chloride by gel filtration using Sephadex G-25 (Pharmacia). Analytical isoelectric focusing of the five resulting pools showed them to contain isoforms 4,5,6,7, and 8. Isoform 4 ran as several bands, indicating that it may have undergone some degradation. The fifth isoform preparation was modified by the addition of a pre-focusing step to the flat bed isoelectric focusing procedure. In this modification, the protein was not added to the ampholyte/urea/gel mixture prior to electrophoresis but was added to the isoelectric focusing apparatus following generation of the pH gradient in the gel bed. Following prefocusing for 75 minutes (1500 volt-hrs) the section of gel bed from 2.25-4.25 cm from the cathode was removed, mixed with the erythropoietin solution, and added back to the gel bed. Following isoelectric focusing, isoforms 10,11,12,13 and 14 were eluted from the gel bed and separated from the ampholytes by ultrafiltration using Centricon-10 (Amicon) devices.
The pre-focusing modification was undertaken to make the ultraviolet absorbance characteristics of the isoform preparations more similar to that of the starting recombinant erythropoietin. This improvement in spectral characteristics can be seen in the ratio of absorbance at 280 and 260 nm for the isolated isoforms. The average ratio of absorbance at 280 nm to that at 260 nm (A280/A260) for isoforms from preparations 2 and 3 (non-prefocused) is 1.36 ± 0.11 while the average
A280/A260 ratio for preparations 5 and 6 (pre-focused) is 1.68 ± 0.20. When isoform #14 is excluded from the calculation, the average A280/A260 ratios are 1.39 ± 0. 11 and 1 .74 ± 0.09 for preparations 2 & 3 and 5 & 6, respectively. (Isoform 14 may have the most atypical spectrum because it is present in the smallest amounts and is thus more subject to interferences by trace contamination by ampholyte components or because it is nearest to the electrode during the flat bed isoelectric focusing procedure). The average A280/A260 ratio for recombinant erythropoietin prepared according to
Example 2 of Lai et al. (modified as described earlier by using Q-Sepharose as the anion exchange resin) is 1.91 ± 0.04.
As described above, the starting material for isoform preparation #6 was a recombinant erythropoietin preparation containing isoforms 4-13. The ampholytes were pre-focused in the Rotofor apparatus as per the fourth preparation. Ampholyte fractions having measured pHs of between 3.7 and 4.8 were used for the flat bed isoelectric focusing. The flat bed was pre-focused as in run #5 and isoforms 9,10,11,12 and 13 were obtained after ultrafiltration (Centricon-10) to remove carrier ampholytes.
Example 2: Sialic Acid Content of Recombinant
Erythropoietin Isoforms
The isoforms isolated as described in Example 1 and erythropoietin purified according to procedures described in Lai et al., supra (mixture of isoforms 9 to 14) are buffer exchanged into 0.10-0.15 M sodium chloride and analyzed for sialic acid content by a modification of the procedure of Jourdian et al.
J. Biol. Chem. 246, 430 (1971). The sialic acid residues are cleaved from the glycoproteins by hydrolysis with 0.35 M sulfuric acid at 80°C for 30 minutes and the solutions are neutralized with sodium hydroxide prior to analysis. In order to estimate the amount of
erythropoietin protein present, a Bradford protein assay (Bradford Anal. Biochem. 72, 248 (1976)) using
recombinant erythropoietin having the amino acid sequence of human erythropoietin as standard is
performed using the assay reagents and the micro-method procedure supplied by Bio-Rad. The results, expressed as moles of sialic acids per mole of erythropoietin, are shown in Table 1. Isoforms are designated according to the number of sialic acids per molecule and range from least acidic (Isoform 9) to most acidic (Isoform 13). Isoforms 9-13 are shown in gel lanes 6-10 of Figure 1. Quantities of Isoform 14 are insufficient to accurately measure the sialic acid content. The sialic acid content of this isoform is inferred from its migration on IEF gels relative to other isoforms. The sialic acid content of isoforms 5-8 (preparation #4) has not been measured but is likewise inferred from their migration on IEF gels.
TABLE 1
ERYTHROPOIETIN MOLES SIALIC ACID/ ISOFORM MOLE ERYTHROPOIETIN
Isoform 13 12.9 ± 0.5
Isoform 12 11.8 ± 0.2
Isoform 11 11.0 ± 0.2
Isoform 10 9.8 + 0.3
Isoform 9 8.9 ± 0.6
Isoform Mixture (9-14) 11.3 ± 0.2
Example 3: Activity of Recombinant Erythropoietin
Isoforms
The isoforms isolated as described in Example 1 are assayed by absorbance at 280 nm, by
Bradford protein assay and by RIA for erythropoietin to determine the amount of recombinant erythropoietin present. The exhypoxic polycythemic mouse bioassay (Cotes et al. Nature 191, 1065 (1961)) is used to determine the relative in vivo biological activity.
Quantitation of the amount of erythropoietin protein present using a radioimmunoassay for erythropoietin produced results having higher relative in vivo specific activity for certain isoforms because of an apparent decreased immunoreactivity of isoforms containing large amounts of sialic acid leading to an underestimation of the erythropoietin concentration and thus an
overestimation of the relative in vivo specific activity for the most negative isoforms. Mouse bioassay
determinations, expressed as units/ml, are divided by the corresponding protein concentrations to give in vivo specific activities expressed as units/mg erythropoietin polypeptide. These specific activities are shown in Table 2.
In Table 2, "n" is the number of independent isoform preparations which contribute to the specific activity value. In most cases several in vivo assays were performed on each isoform preparation. The same in vivo data contribute to the specific activity
calculations for all three columns, units/mg
erythropoietin polypeptide was determined by the
absorbance at 280 nm, from radioimmunoassay potencies, or from Bradford protein assay results. Purified recombinant erythropoietin containing isoforms 9-14 was used as the standard in the Bradford protein assay. "n" may be less for the calculation made using the Bradford protein assay as some preparations were no longer available at the time the Bradford assays were
performed.
Erythropoietin purified according to the procedures described in Lai et al., supra and containing a mixture of isoforms 9 to 14 is used as a standard for the RIAs and in vivo assays.
The relative specific activities expressed as units/mg erythropoietin polypeptide can be converted to units/A280 by multiplying by 0.807 mg erythropoietin polypeptide/A280. The conversion factor is derived by multiplying the extinction coefficient of erythropoietin (1.345 mg/A280) by the protein content of the
erythropoietin glycoprotein (about 60% by weight, Davis et al. Biochemistry 26, 2633 (1987)) to obtain mg erythropoietin polypeptide/A280 (i.e., 1.345 mg erythropoietin/A280 × 0.60 mg polypeptide/mg
erythropoietin = 0.807 mg polypeptide/A280). In addition, specific activities expressed as units/mg erythropoietin polypeptide can be multiplied by the factor 0.60 mg polypeptide/mg erythropoietin
glycoprotein to give specific activities expressed as units/mg erythropoietin glycoprotein.
Figure imgf000033_0001
The data in Table 2 are also presented
graphically in Figures 2A, 2B and 2C. These data show that the relative in vivo activity of erythropoietin increases as a function of sialic acid content up until isoform #11. Isoforms 11-14 have essentially the same relative in vivo bioactivity. (This is most apparent when the concentration of isoform 14 is expressed using the Bradford assay value. The Bradford value may be more accurate for isoform 14 because of the generally low levels obtained and the resulting difficulty in
determination by A280 and the most apparent decreased reactivity in the RIA of very negative forms discussed previously). The greater relative in vivo specific activity of erythropoietin isoforms having more sialic acid is most likely due to a longer circulating half-life of these forms. Isoforms 9 and 13 were labeled with radioactive iodine (125I) and their rate of clearance in rats was determined. The half-life in circulation was significantly longer for isoform 13 than for isoform 9.
Example 4: Selection of Recombinant Erythropoietin
Isoform Mixtures by O-Sepharose Chromatography
Cell conditioned media from the production of recombinant erythropoietin according to the procedures described in Lin, supra are concentrated and diafiltered against 10 mM Tris, pH 7.2. Protein concentration is determined by the Bradford microprotein assay using bovine serum albumin as a standard. 19.6 ml of the solution containing 40 mg of total protein is made 20 μM in CUSO4, filtered through a 0.45 micron cutoff filter and loaded onto a 4 ml bed volume (1.05 cm height × 2.2 cm diameter) column packed with Q Sepharose Fast Flow (Pharmacia) which has been equilibrated with 10 mM Tris, pH 6.8 to 7.0 at 4°C. After sample application, the column is washed with two column volumes of the same buffer. The column flow rate is about 1 ml/min. Six separate columns are set up using this procedure to select defined erythropoietin isoform mixtures.
Columns are washed with 6 to 9 column volumes of a low pH buffer consisting of: Column #1, 150 mM acetic acid, 1 mM glycine, 20 μM CUSO4, 6 M urea adjusted to pH 4.7 with NaOH; Column #2, 200 mM acetic acid, 1 mM glycine, 20 μM CuSO4, 6 M urea adjusted to pH
4.7 with NaOH; Column #3, 250 mM acetic acid, 1 mM glycine, 20 μM CUSO4, 6 M urea adjusted to pH 4.7 with NaOH; Column #4, 300 mM acetic acid, 1 mM glycine, 20 μM CUSO4, 6 M urea adjusted to pH 4.7 with NaOH; Column #5, 150 mM acetic acid, 1 mM glycine, 20 μM CuSO4, 6 M urea; Column #6, 300 mM acetic acid, 1 mM glycine, 20 μM
CUSO4, 6 M urea. The pH of the columns is increased to approximately pH 7 by washing each one with 8 to 11 column volumes of 10 mM Tris-HCl, 55 mM NaCI, 20 μM
CUSO4, pH 7. The defined erythropoietin isoform
mixtures are eluted from the columns by washing with 10 mM Tris-HCl, 140 mM NaCI, 20 μM CuSO4, pH 7.0.
The eluted isoform pools from each column are concentrated and solvent exchanged into water using an Amicon Centricon-10 microconcentrator. The results of analytical isoelectric focusing of these concentrated pools are shown in Figure 3. Gel lanes 1-6 represent defined erythropoietin isoform mixtures eluted from column 1-6, respectively. The "isoform mixture" shown in the far right gel lane of Figure 3 represents cell media which is applied to a Q-Sepharose column as described above, the column is washed with 5 mM acetic acid, 1 mM glycine, 20 μM CuSO4, 6M urea, and the erythropoietin isoform mixture is eluted from the column using the procedures described above. This eluted mixture of isoforms is further purified according to the procedures described in Lai et al., supra prior to analytical isoelectric focusing. Example 5: Fractionation of Recombinant Erythropoietin Isoforms Using a Low pH Gradient on O-Sepharose In another procedure, erythropoietin isoforms are separated using a gradient of decreasing pH and increasing ionic strength. The concentrated diafiltered erythropoietin containing media is loaded to a column of Q-Sepharose at a ratio of approximately 40 mg total protein/mL gel. The column is then washed with
approximately two column volumes of 10 mM Tris HCI, pH 7.0 and then approximately 10 column volumes of 2 mM acetic acid/1 mM glycine/20 μM CUSO4/6 M urea (pH approximately 4.8) to remove contaminating proteins and erythropoietin isoforms containing less than
approximately 7 sialic acid residues. Isoforms
containing from approximately 8 to approximately 12 sialic acids are eluted from the column using a gradient starting at approximately 2 mM acetic acid in 6 M urea/1 mM glycine/20 μM CUSO4 and running to 40 mM acetic acid/6 M urea/1 mM glycine/20 μM CUSO4
(pH approximately 4). The total volume of the gradient is approximately 40 column volumes and fractions of approximately one column volume each are collected into vessels containing a volume of Tris buffer sufficient to bring the pH into the range of 6-8.5 so as to avoid long term exposure of the collected fractions to low pH.
Aliquots of the fractions are subjected to analytical isoelectric focusing to monitor the separation.
Figure 4 shows the separation of isoforms 8-11 which may be achieved by this procedure. Isoforms 12-14 which remain bound to the column at the end of the gradient are eluted by washing with a buffer consisting of 10 mM TrisHCl, 140 mM NaCI, 20 mM CuSO4 (pH 7.0). The isoforms (separated during the gradient or eluted by the sodium chloride solution) are freed of contaminating proteins by reverse phase chromatography followed by gel
filtration chromatography as described in Example 2 of Lai et al. Example 6: Construction of Human Erythropoietin Analogs
The locations of existing carbohydrate attachment sites within the human erythropoietin amino acid sequence are shown in Figure 5 (SEQ ID. NO: 26). Procedures for generating additional glycosylation sites for erythropoietin are summarized in Figures 6A-C and described below.
The following oligonucleotide primers were synthesized for use in in vitro mutagenesis:
[Asn4, Ser6] EPO: 5' CGCCCACCAAACCTCAGCTGTGACAGCCGA 3'
(SEQ ID NO: 1)
[Asn9, Ser11] EPO: 5' ATCTGTACAACCGAAGCCTGGAGAGGT 3'
(SEQ ID. NO: 2)
[Asn69] EPO: 5' GGGCCTGGCCAACCTGTCGGAAG 3' (SEQ ID. NO: 3)
[Asn124] EPO: 5' TCCCCTCCAGATAATGCCTCAGCTGC 3' (SEQ ID. NO: 4) [Asn125, ser->-27] EPO: 5' CAGATGCGAACTCATCTGCTCCAC 3'
(SEQ ID. NO: 5)
[Asn163, Ser165] EPO: 5' AGGCCTGCAGGAATGGGAGCAGATGACCAGGTG 3' (SEQ ID. NO: 6)
[Thr125] EPO: 5' TCCAGATGCGACCTCAGCTGCTC 3' (SEQ ID. NO: 7)
[Pro124, Thr125] EPO: 5' CCTCCAGATCCGACCTCAGCTGC 3'
(SEQ ID. NO: 8) The underlined codons show the mismatched regions where the amino acids indicated in brackets replace the wild-type amino acids.
[Asn4, Ser6] EPO was constructed to add an
N-glycosylation site at Asn 4. [Asn9, Ser11] EPO was constructed to add an N-glycosylation site at Asn 9.
[Asn69] EPO was constructed to add an N-glycosylation site at Asn 69. [Asn125, Ser127] EPO was constructed to add an N-glycosylation site at Asn 125. [Thr125] EPO and [Pro124, Thr125] EPO were constructed to add an
O-glycosylation site at Thr 125.
The following oligonucleotide primers were synthesized for use in in vitro mutagenesis:
[Asn69, Thr71] EPO: 5' GGGCCTGGCCAACCTGACAGAAGCTGTC 3'
(SEQ ID. NO: 9)
[Ser68, Asn69, Thr71] EPO:
5' CAGGGCCTGTCCAACCTGACAGAAGCTGTC 3'
(SEQ ID. NO: 10) [Asn125, Thr127] EPO: 5' CAGATGCGAACTCAACGGCTCCAC 3'
(SEQ ID. NO: 11)
[Asn125, Thr127, Thr131] EPO:
5ΑTGCGAACTCAACGGCTCCACTCACAACAATCACT 3'
(SEQ ID. NO: 12)
[Pro124, Asn125, Ser127] EPO:
5' CCAGATCCAAATTCATCTGCTCCACTC 3'
(SEQ ID. NO: 13)
[Pro124 , Asn125, Thr127] EPO :
5 ' CCAGATCCAAATTCAACAGCTCCACTC 3 '
(SEQ ID . NO : 14 ) [Thr125, Thr126] EPO: 5' CCAGATGCGACAACAGCTGCTCCA 3'
(SEQ ID. NO: 15)
[Proi24, Thr125, Thr126, Thr131] EPO:
Starting from [Pro124, Thr125] EPO cDNA, the oligonucleotide primer 5' AGATCCGACCACCGCTGCTCCAC 3' (SEQ ID. NO: 16) is used to generate [Pro124, Thr125, Thr126] EPO. The oligonucleotide primer
5'TGCTCCACTCACAACAATCACTG 3' (SEQ ID. NO: 17) is then used to generate [Pro124, Thr125, Thr126, Thr131] EPO. [Asn69, Thr71] EPO and [Ser68, Asn69, Thr71]
EPO are constructed to add an N-glycosylation site at Asn 69 and to enhance N-glycosylation at that site. [Asn125, Thr127] EPO, [Asn125, Thr127, Thr131] EPO,
[Pro124, Asn125, Ser127] EPO and [Pro124, Asn125, Thr127] EPO are constructed to add an N-glycosylation site at Asn 125 and to increase glycosylation at that site . [Thr125, Thr126] EPO and [Pro124, Thr125, Thr126, Ser131] EPO are constructed to add an O-glycosylation site at Thr 125 and to increase glycosylation at that site.
The source of erythropoietin DNA for in vitro mutagenesis was plasmid Hul3, a human erythropoietin cDNA clone in pUC 8 (Law et al. Proc Natl. Acad. Sci. 83. 6920 (1986)). Plasmid DNA derived from Hu13 was digested with BstEII and BglII restriction enzymes, the resulting DNA fragments were subjected to agarose gel electrophoresis, and the 810 base pair (bp)
erythropoietin DNA fragment was isolated from the gel using a GeneClean™ kit and procedures supplied by the manufacturer (BIO 101, Inc.). Plasmid pBRgHuEPO contains the erythropoietin genomic gene as a BamHI fragment inserted into a derivative of pBR322, as described in commonly owned Lin patent, supra.
pBRgHuEPO was also digested with BstEII and BglII and the 6517 bp vector fragment was recovered. Ligation of the two fragments results in IGT1. To construct pEC-1, pDSVL (described in commonly owned Lin patent, supra, and shown in Figure 5B) was digested with BamHI and the isolated 2.8 kilobase (kb) BamHI fragment from IGT1 containing erythropoietin cDNA was ligated into it.
In order to generate single-stranded DNA for in vitro mutagenesis, pEC-1 was digested with BamHI and BglII and the 820 bp erythropoietin cDNA fragment was isolated. It was ligated into the BamHI site of m13mp18 to give m13-EC-1. Single stranded DNA was recovered from supernatants of E. coli strain RZ1032 infected by m13-EC-1 as described by Kunkel et al. Methods in
Enzymol. 154. 367 (1987) and Messing, Methods in
Enzymol. 101. 20 (1983). For in vitro mutagenesis approximately 1 μg of single-stranded DNA and 0.2 pmole of one of the synthetic primers described above were mixed with 6 μl of buffer (250 mM Tris pH 7.8, 50 mM
MgCl2, and 50 mM dithiothreitol). For annealing of the primer to the template, the reaction volume was adjusted to 10 μl with water, the mixture was heated to 65°C for
5 minutes and then allowed to cool to room temperature. For the elongation reaction 2.5 μl of each of dTTP, dATP, dGTP, dCTP and ATP (all at 10 μM) were added, followed by 1 μl (1 unit) of E. coli DNA polymerase (Klenow fragment) and 1 μl (1 unit) of T4 DNA ligase.
The mixture was then incubated overnight at 14°C and used to transform E. coli JM 109 (Yanisch-Perron et al. Gene 33. 103 (1985)) as described (Messing, supra).
To identify mutant clones by differential hybridization, plaques on nutrient agar were transferred to Gene Screen filters (New England Nuclear). The filters were dried under a heat lamp and then incubated for one hour in 6x SSC containing 1% SDS at 60°C . For the hybridization, the oligonucleotide primer above (8 pmoles) was end-labeled with T4 polynucleotide kinase and γ 32P-iabeled ATP and incubated with the filters overnight in 6x SSC, 0.5% SDS and 100 mg/ml salmon sperm DNA at 37°C for the [Asn124] mutation, 55°C for the
[Asn4, Ser6] mutation, 65°C for the [Thr125] and the
[Pro124, Thr125] mutations, and 70°C for the [Asn9, Ser11] and [Asn163, Ser165] mutations. The next day, the filters were washed three times with 6x SSC at room temperature and subjected to autoradiography. If necessary, the filters were then washed with 6x SSC at increasing temperatures until little or no hybridization was detected to plaques having the wild-type
erythropoietin cDNA sequence. Clones that gave positive hybridization signals under these conditions were identified and retransfected into JM109 to isolate a pure clone. Dideoxy chain termination sequence analysis indicated that the mutations to asparagine, serine threonine and proline residues were present.
Double stranded ml3 EC-1 DNAs carrying the
[Asn4, Ser6], [Asn9, Ser11], [Asn69], [Asn124], [Asn125], [Ser127], [Asn163, Ser165] [Thr125], and [Pro124, Thr125] changes were recovered from JM109 transfected cells by the boiling method (Holmes et al. Anal. Biochem 117, 193 (1981)). The DNAs were digested with BstEII and XhoII and the 810 bp erythropoietin DNA fragments were
isolated. pEC-1 was digested with BstEII followed by a partial digestion with BglII and the 5' termini of the resulting fragments are dephosphorylated with bacterial alkaline phosphatase in 10 mM Tris, pH 8 at 60°C for 60 minutes. The 7 kb vector fragment lacking the 810 bp BstEII-BglII fragment was isolated and ligated to the erythropoietin fragments above. The resulting plasmids (designated pEC-X where X is the analog number) contain DNA encoding erythropoietin analogs having altered amino acid residues at the indicated positions.
Alternatively, an analog of erythropoietin (pEC34) was constructed by in vitro mutagenesis that deleted amino acid residues 41-55. This resulted in a smaller (775 bp) EPO containing BstEII-BglII fragment. The fragment was inserted into pECl as described above. For cloning analogs of erythropoietin, pEC34 was digested with BstEII, partially digested with BglII, dephosphorylated and the vector isolated as described above. The 7kb vector fragment was then ligated to erythropoietin fragments as described above. Cloning with pEC34 allows easy discrimination between
recombinants and simple reclosures. Reclosures result in a smaller BstEII-BglII fragment than analogs and these can be easily differentiated on agarose gels.
These general procedures were used to construct the erythropoietin analogs shown in Tables 3, 4 and 5. The DNA sequence changes for each of the analogs are shown; otherwise the oligonucleotide primers used for mutagenesis had sequences complimentary to those of human erythropoietin.
TABLE 3
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR N-LINKED CARBOHYDRATE CHAINS
Amino Acid Sequence Analog No. Substitution Changes
N1 Asn4Ser6 CGC→AAC
ATC→AGC N2 Asn9ser11 AGC→AAC
GTC→AGC N3 Asn19Thr21 GCC→AAC
GAG→ACG N4 Asn30Thr32 GCT→AAT
CAC→ACG N5 Asn42 CCA→AAT
N6 Ser42Asn43Thr45 CCA→TCA
GAC→AAC
AAA→ACA N7 Ser49 TAT→TCT
N8 Asn51Thr53 TGG→AAT
AGG→ACG N9 Asn57Thr59 GGG→AAC
CAG→ACG N10 Asn69 CTG→AAC
N11 Asn69Thr71 CTG→AAC
TCG→ACA N12 Ser68Asn69Thr71 N11 plus
GCC→TCC N13 Asn88Thr90 TGG→AAT
CCC→ACC N14 Ser87Asn88Thr90 N13 plus
CCG→TCG N15 Ser87Asn88Thr90Thr92 N14 plus
CAG→ACG TABLE 3 (CONT'D)
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR N-LINKED CARBOHYDRATE CHAINS
Analog Amino Acid Sequence No. Substitution Changes
N16 Ser87Asn88Thr90Ala162 N14 plus
AGG→GCG
N17 Asn88Ser90 TGG→AAT
CCC→TCC
N18 Val87Asn88Thr90 CCG→GTG
TGG→AAT
CCC→ACC
N19 Ser87Asn88Gly89Thr90 CCG→TCG
TGG→AAT
GAG→GGG
CCC→ACC
N20 Asn89Ile90Thr91 GAG→AAC
CCC→ATC
CTG→ACG
N21 Ser87Asn89lle90Thr91 N20 plus
CCG→TCG N22 Asn113Thr115 GGA→AAC
CAG→ACG N23 Asn118Val121 GCC→AAC
CCT→GTT N24 Asn121Ser122Thr123 CCT→AAT
CCA→TCA GAT→ACT
N25 Asn124 GCG→AAT N26 Ser122Asn124 CCA→TCA
GCG→AAC
N27 Asn125Ser127 GCC→AAC
GCT→TCT
N28 Asn125Thr127 GCC→AAC
GCT→ACG TABLE 3 (CONT'D)
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR N-LINKED CARBOHYDRATE CHAINS
Analog Amino Acid Sequence No. Substitution Changes
N29 Leu121Ser122Asn125Thr127 N28 plus
CCT→CTT
CCA→TCA N30 Thr120Gly122Leu123Asn125Thr127 N28 plus
TCC→ACC
CGC→GGG
GAT→CTC
N31 Asn126Ser128 TCA→AAT
GCT→TCT
N32 Asn126Thr128Val129 TCA→AAT
GCT→ACT CCA→GTA
N33 Leu121Ser122Asn126Thr128Val129 N32 plus
CCT→CTT
CCA→TCA N34 Thr121Gly123Ser125Asn126Thr128Val 1 29 N32 plus
CCT→ACG
GAT→GGG
GCC→TCC N35 Ser129Asn130 CCA→AGC
CTC→AAC N36 Asn132 ACA→AAC
N37 Asn134Thr136 ACT→AAT
GAC→ACC N38 Asn 1 35 GCT→AAT
N39 Asn 136Thr 1 38 GAC→AAC
TTC→ACC N40 Asn137Thr139 ACT→AAT
CGC→ACC N41 Asn 138Thr140 TTC→AAC
AAA→ACA TABLE 3 (CONT'D)
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR N-LINKED CARBOHYDRATE CHAINS
Analog Amino Acid Sequence No . Substitution Changes
N42 Asn144 GTC→AAC
N43 Ser143Thr149Val150 TTC→TCC
CTC→ACC CGG→GTG
N44 Gly148Thr149 TTC→GGC
CTC→ACC
N45 Asn155 CTG→AAT
N46 Asn163Ser165 ACA→AAT
N47 Asn30Thr32Val87Asn88Thr90 N4 and N18
N48 Asn69Thr71Ser87Asn88Thr90 N11 and N14
TABLE 4
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR O-LINKED CARBOHYDRATE CHAINS
Amino Acid Sequence
Analog No . Substitution Change
01 Ser6 ATC→AGC
02 Ser7 TGT→TCC
03 Ser8 GAC→AGC
04 Ser11 GTC→TCT
05 Ser18 GAG→TCG
06 Ser23 GAG→TCG
07 Ser29 TGT→AGC
08 Ser29 TGT→TCT
09 Ser30 GCT→TCT
O10 Ser33 TGC→TCA
O11 Ser37 GAG→TCG
012 Ser49 TAT→TCT
013 Ser61 GTA→TCA
014 Ser63 GTC→TCC
015 Ser67 CTG→TCG
016 Ser68 GCC→TCC
017 Ser70 CTG→TCG
018 Ser73 GCT→TCT
019 Ser74 GTC→TCT
020 Ser75 CTG→TCG
021 Ser79 GCC→TCC
022 Ser81 TTG→TCG
023 Ser86 CAG→TCG
024 Ser87 CCG→TCG
025 Ser93 CTG→TCG
026 Ser98 GCC→TCC
027 Ser99 GTC→TCC
028 Ser102 CTT→TCT
029 Ser103 CGC→AGT TABLE 4 (CONT'D)
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR O-LINKED CARBOHYDRATE CHAINS
Amino Acid Sequence
Analog No . Substitution Change
030 Ser105 CTC→AGC
031 Ser109 CTT→TCT
032 Ser111 GCT→TCT
033 Ser112 CTG→TCG
034 Ser114 GCC→TCC
035 Ser128 GCT→TCT
036 Ser159 TCG→GAG
037 Ser1 61 TGC→TCC
038 Pro125Ser127 GCC→CCC
GCT→TCT
039 Thr62 GAA→ACA
040 Thr64 TGG→ACG
041 Thr65 CAG→ACG
042 Thr88 TGG→ACG
043 Thr90 CCC→ACC
044 Thr92 CAG→ACG
045 Thr100 AGT→ACT
046 Thr110 CGG→ACG
047 Thr1 15 CAG→ACG
048 Thr123 GAT→ACT
049 Thr124 GCG→ACG
050 Thr125 GCC→ACC
051 Thr125Ser127 GCC→ACC
GCT→TCT
052 Thr125Thr126 GCC→ACA
TCA→ACA
053 Thr126 TCA→ACC
054 Thr127 GCT→ACT
055 Thr 130 CTC→ACC TABLE 4 (CONT'D)
ERYTHROPOIETIN ANALOGS HAVING SITES
FOR O-LINKED CARBOHYDRATE CHAINS
Amino Acid Sequence
Analog No. Substitution Change
056 Thr131 CGA→ACA
057 Thr136 GAC→ACC
058 Thr140 AAA→ACA
059 Pro124Thr125 GCG→CCG
GCC→ACC
060 Pro124Thr125Thr126 059 plus
TCA→ACC
061 Pro124Thr125Thr126Thr131 060 plus
CGA→ACA
062 HCG C-terminal extension
TABLE 5
ERYHROPOIETIN ANALOGS HAVING SITES
FOR N- AND O-LINKED CARBOHYDRATE CHAINS
Analog Amino Acid Sequence
No. Substitution Change
Ser87Asn88Thr90 N14 and 062
NO1 HCG C-terminal extension
Asn30Thr32Val87Asn88Thr90 N47 and 062 NO2 HCG C-terminal extentsion
Plasmids designated pDEC-X (where X is the analog number) were constructed by inserting erythropoietin cDNA into pDECΔ, which is a derivative of plasmid pDSα2. The expression vector pDSα2 is generally described in
PCT Application No. WO 90/14363. pDECΔ was derived from pDSα2 by the following steps:
(1) The HindIII site of pDSα2 was deleted by digesting pDSα2 DNA with HindIII, treating the HindIII cohesive ends with E. coli DNA Polymerase (Klenow fragment) and dNTPs, and religating the blunt-ended vector. The resulting plasmid was pDSα2ΔH.
(2) pDSα2ΔH was digested with Sail and a synthetic oligonucleotide having an SV40 splice signal with a Sail linker attached to the 3' end of the splice signal was ligated to it. The synthetic oligonucleotide had the following sequence (SEQ ID. NO: 18) :
5' TCGAGGAACTGAAAAACCAGAAAGTTAACTGGTAAGTTTAGT CTTTTTGTCTTTTATTTCAGGTCCCGGATCCGGTGGTGGTGCAAATCA AAGAACTGCTCCTCAGTGGATGTTGCCTTTACTTCTAGGCCTGTACGG AAGTGTTACTTCTGCTCTAAAAGCTGCTGCAACAAGCTGGTCGACC 3' The resulting plasmid was pDSα2ΔH splice.
3) pDSα2ΔH splice was digested with SalI and blunt-ended by treating the cohesive ends with T4 DNA polymerase and dNTPs. An 820 bp. BamHI-BglII human erythropoietin cDNA fragment was blunt-ended by the same method and ligated to the plasmid. The resulting plasmid was pDEC-1.
4) pDEC was digested with KpnI and PvuII and blunt-ended by treating the cohesive ends with mung bean nuclease. The plasmid was religated to delete the excised KpnI-PvuII fragment resulting in the plasmid pDECΔ.
pDEC-X plasmids were made from pDECΔ by complete digestion with BstEII followed by a partial digestion with BglII. The vector fragment lacking erythropoietin coding sequences was isolated and ligated to the 810 bp BstEII-BglII fragments containing the desired plasmid.
Details of the construction of some analogs having multiple amino acid changes are described below.
Construction of pDEC(N47) and pDEC(N48) pDEC(N47) which contains asn30 thr32 val87 asn88 and thr90 mutations was constructed from pDEC(N18) and pDEC(N4). pDEC(N18) was digested with HindII and BglII and a 445 bp fragment was isolated. pDEC(N4) was digested with BstEII and HindIII and a 377 bp fragment was isolated. These two fragments were ligated into pDECΔ cut with BstEII and BglII as described above resulting in pDEC(N47).
pDEC(N48) which contains asn69 thr71 ser87 asn88 and thr90 mutations was constructed from pDEC(N14) and pDEC(N11). pDEC(N14) was digested with Hindi! and BglII and a 445 bp fragment was isolated. pDEC(Nll) was digested with BstEII and HindII and a 377 bp fragment was isolated. These two fragments were ligated into pDECΔ cut with BstEII and BglII as described above
resulting in pDEC(N48).
Construction of pDEC(062) (HCG-Erythropoietin Fusion) pDEC(062) was assembled from pECl and a 107 base pair StuI-BglII synthetic DNA linker containing the carboxy terminal 28 amino acids from human chorionic gonadotropin (ser-ser-ser-ser-lys-ala-pro-pro-pro-ser-leu-pro-ser-pro-ser-arg-leu-pro-gly-pro-ser-asp-thr-pro-ile-leu-pro-gln) (SEQ ID. NO: 25) (Pierce et al. Ann.
Rev. Biochem. 50, 465 (1981)). The sequence of the linker is as follows:
5' CCTGTAGGACAGGGGACAGATCCTCTTCCTCAAAGGCCCCTCCCCCCAGCCTTC- 3' GGACATCCTGTCCCCTGTCTAGGAGAAGGAGTTTCCGGGGAGGGGGGTCGGAAG-
5 ' CAAGTCCATCCCGACTCCCGGGGCCCTCGGACACCCCGATCCTCCCACAATGA (SEQ ID. NO: 19)
3 ' GTTCAGGTAGGGCTGAGGGCCCCGGGAGCCTGTGGGGCTAGGAGGGTGTTACTCTAG (SEQ ID. NO: 20) pECl was digested with StuI and BglII and the 610bp DNA fragment was isolated. The synthetic linker was phosphorylated with ATP and polynucleotide kinase and ligated with the pECl fragment into pDECΔ previously digested with BstEII and partially digested with BglII as described above. Construction of pDEC(NO1) pDEC(NO1) was assembled from pDEC(062) (HCGEPO) and pDEC(N14) (Ser87Asn88Thr90). pDEC177 was digested with StuI and BglII and the 610bp DNA fragment
containing the Ser87Asn88Thr90 mutations was isolated with gene-clean. pDEC(062) was digested with StuI and BglII and the 107 base pair fragment was isolated. These two DNA fragments were ligated into pDECΔ previously digested with BstEII and partially digested with BglII as described above.
Construction of pDEC(NO2) pDEC(N02) was assembled from pDEC(062) (HCG- EPO) and pDEC(N47) (Asn30Thr32Val87Asn88Thr90). pDEC(N47) was digested with StuI and BglII and the 610bp DNA fragment containing the Asn30Thr32Val87Asn88Thr90
mutations was isolated with GeneClean. pDEC(O62) was digested with StuI and BglII and the 107 base pair fragment was isolated. These two DNA fragments were ligated into pDECΔ previously digested with BstEII and partially digested with BglII as described above. Construction of PDEC(N16) (Ser87Asn88Thr90Ala162) pDEC(N16) was assembled from
pDEC(N14) (Ser87Asn88Thr90) and pDEC258 (Ala162). pDEC258 was constructed using procedures for in vitro
mutagenesis described above and changing the AGG codon to GCG at position 162. pDEC(N14) was digested with StuI and BglII and the 610bp DNA fragment containing the Ser87Asn88Thr90 mutations was isolated with GeneClean™. pDEC258 was digested with StuI and BglII and a 210 base pair fragment was isolated. These two DNA fragments were ligated into pDECΔ previously digested with BstEII and partially digested with BglII as described above.
Construction of pDEC(R1), (R2) and (R3)
To remove glycosylation sites from pDEC(N14), ml3-EPO(N14) containing ser87 asn88 and thr90 mutations was subjected to in vitro mutagenesis as described above using the following primers:
5' GGAGGCCGAGCAGATCACGACGG3' GLN24
(SEQ ID. NO: 21)
5' CTTGAATGAGCAGATCACTGTCC3 ' GLN38
(SEQ ID. NO: 22)
5' CTGTTGGTCCAGTCTTCCCAG3 ' GLN83
(SEQ ID. NO: 23)
The resulting plasmids were designated pDEC(R1) (gln24 ser87 asn88 thr90) pDEC(R2) (gln38 ser87 asn88 thr90) and pDEC(R3) (gln83 ser87 asn88 thr90). m13EC-1 was also subjected to in vitro mutagenesis with the above
oligonucleotide primers resulting in pECIO (gln24) and pEC8 (gln38). pEC9 (gln83) was constructed using the primer
5' CCTGTTGGTCCAGTCTTCCCAGC3 ' GLN83
(SEQ ID. NO: 24) cDNA clones of human erythropoietin and analogs corresponding to [Asn4, Ser6] EPO, [Asn9, Ser11] EPO, [Asn69] EPO, [Asn124] EPO, [Asn125, Ser127] EPO,
[Asn163, Ser165] EPO, [Thr125] EPO and [Pro124, Thr125] EPO and cDNA clones of analogs described in Tables 3,4 and 5, were transferred into COS-1 cells (ATCC No. CRL-1650) by electroporation. COS-1 cells were harvested from semi-confluent dishes, washed with medium
(Dulbecco's modified essential medium containing 5% fetal calf serum and 1% L-glutamine/penicillin/
streptomycin (Irvine Scientific) ) and resuspended at 4 × 106 cells/ml. One ml of cells was transferred to an electroporation cuvette (Bio-Rad) and electroporated with a Bio-Rad Gene Pulser at 25 μFarads and 1600 volts in the presence of 100 to 200 μg of carrier DNA and 2 to 20 μg of plasmid DNA encoding the erythropoietin analog. The electroporated cells were plated at 2 × 106 cells per 60 mm tissue culture dish in 5 ml of medium. Two to four hours after plating the medium was replaced with 5 ml of fresh medium. The conditioned medium was
collected 3 to 5 days after electroporation.
Example 7: Characterization of Erythropoietin Analogs A. Determination of Carbohydrate Addition
A volume of supernatant containing 5-20 units from COS cells transfected with erythropoietin analog cDNAs as described in Example 6 was immunoprecipitated overnight at room temperature with a rabbit antierythropoietin polyclonal antibody. 20-80 μl of 1:1 Protein A-Sepharose in phosphate buffered saline (PBS) was added to the immunoprecipitate and allowed to incubate for one hour at room temperature. The samples were centrifuged, washed with PBS and, where indicated, the pellet was treated with N-glycanase to remove
N-linked carbohydrate chains. The samples were analyzed by 15% SDS-polyacrylamide gel electrophoresis,
transferred to nitrocellulose and subjected to Western analysis as described (Burnette et al. Anal. Biochem. 112, 195-203 (1981); Elliott et al. Gene 79 , 167-180 (1989)) using a mixture of mouse anti-erythropoietin monoclonal antibodies. One such antibody, 9G8A, is described in Elliott et al. (1989) Blood 74, Supp. 1, A. 1228. Analysis of COS cell supernatants transfected with [Asn69] EPO and [Asn125, Ser127] EPO cDNA revealed increased protein size compared to human sequence erythropoietin. This increased size is indicative of an additional N-linked carbohydrate chain (Figure 7).
Treatment of supernatants from COS cells transfected with [Thr125] EPO and [Pro124, Thr125] EPO cDNA with N-glycanase revealed an increased protein size compared to human sequence erythropoietin. This increased size is indicative of an additional O-linked carbohydrate chain (Figure 8). Western blot analysis of other selected analogs is shown in Figure 10.
To determine the number of N-linked carbohydrate chains attached to EPO, partial N-glycanase digestions were performed. Analogs or rHuEPO were expressed in CHO cells and conditioned serum free medium was collected. Tubes contained 40 units of EPO (volume adjusted to 15 μl with H2O) To each tube was added 10 μl
0.5% SDS and each sample was boiled for 3 minutes. Then the following components were added, 10.8 μl 0.5M NaPθ4 pH8.6, 5μl 7.5% nonidet P40 and 1.5 μl of 250 unit/ml N-glycanase (Genzyme). Samples were incubated for the indicated times at 37°C. The reaction was stopped by the addition of SDS-PAGE sample buffer (see above) and then subjected to SDS-PAGE western analysis (10% acrylamide) using an anti-EPO polyclonal antibody and a anti-rabbit Vectastain™ kit (Vector laboratories) with
4-chloronaphthol as substrate. An analysis of N-linked chains using this method is shown in Figure 11 for human erythropoietin and analog N14.
B. Assays for erythropoietin analog activity
RIAs were performed according to Egrie et al., supra. EIAs were performed with CLINIGEN™ EIA kit (R and D Systems) using procedures supplied by the manufacturer. The in vivo biological activity of erythropoietin analogs was determined on supernatants from CHO cells expressing an erythropoietin analog or on purified erythropoietin obtained from CHO cell
conditioned medium as described below using the
exhypoxic polycythemic mouse bioassay (Cotes et al., supra).
In vitro erythropoietin activity was determined by the erythroid colony forming assay as described by Iscove et al. J. Cell Physiol. 83, 309-320
(1974) with modifications. The mononucleated cells from human bone marrow cells were partially purified on a ficoll-paque cushion and washed in Iscove medium before plating to remove the adherent cells. The culture medium contained 0.9% methyl cellulose and did not include any bovine serum albumin. The erythroid
colonies were scored after 8 to 10 days of culture.
The erythropoietin analogs transfected and expressed in COS cells as described in Example 6 were analyzed in crude COS cell supernatants using RIA, EIA and an erythroid colony forming assay. Purified human sequence erythropoietin has an in vitro activity that was comparable to the RIA activity as determined by the above-mentioned assays. The analogs [Asn69] EPO,
(Thr125) EPO and [Pro124, Thr125] EPO exhibited an
in vitro activity that was comparable to the RIA
activity and gave evidence of having additional
carbohydrate chains (as determined in Section A) .
[Thr125] EPO and analogs N4, N11, N14, N16, N18, N47, N48, 062, NO1 and NO2 were analyzed further by
transfecting a cDNA clone encoding the erythropoietin analog into CHO cells, and subjecting the CHO cell supernatants to RIA or EIA and in vivo biological assays. The results are shown in Table 6. in vivo activities for analogs R1, R2 and R3 expressed in CHO cell supernatants are shown in Table 7. TABLE 6
ERYTHROPOIETIN ANALOGS HAVING ADDITIONAL CARBOHYDRATE CHAINS
In Vivob
N-Linkeda O-Linked Activity
EPO Sequence Chains Chains RIA or EIA
Human 3c 1d 0.6
Asn30Thr32 3 to 4c n.t. 1.1
Asn51Thr53 4 n.t. n.t.
Asn57Thr59 4 n.t. n.t.
Asn69 4 reducede n.t.
Asn69Thr71 4 n.t. 0.25-0.7
Ser68Asn69Thr71 4 n.t. n.t.
Val87Asn88Thr90 4c n.t. 1.8
Ser87Asn88Thr90 3 to 4c normal 1.0
Ser87Asn88Gly89Thr90 4 n.t. n.t.
Ser87Asn88Thr90Thr92 4 reducede n.t.
Asn89Ile90Thr91 4 n.t. n.t.
Ser87Asn89Ile90Thr91 4 n.t. n.t.
Ser87Asn88Thr90Ala162 4 n.t. 1.8
Asn136Thr138 3 to 4 n.t. n.t.
Asn138Thr140 3 to 4 n.t. n.t.
Asn69Thr71Ser87Asn88Thr90 4 to 5 n.t. 0.025-0.25
Asn30Thr32Val87Asn88Thr90 4 to 5c normal 1.8
Thr123 3 1 and 2f n.t.
Thr125 3 1 and 2f 0.7
Pro124Thr125 3 1 and 2f n.t.
HCG C-terminal extension 3 at least 3g 1.0-1.3
Ser87Asn88Thr90
HCG extension 4 at least 3g 1.5
Asn30Thr32Val87Asn88Thr90
HCG extension 4 to 5 at least 3g 0.8 TABLE 6 FOOTNOTES
aThe number of additional N-linked chains was estimated based upon the mobility of the analog polypeptides in SDS gels as described in Example 7A.
bRatio of in vivo activity of analog to amount of
erythropoietin analog. Activity measurements were done on CHO cell supernatants of analogs using the mouse
polycythemic bioassay. Quantities of erythropoietin analogs in CHO cell supernatants were determined by RIA or EIA as described in the text.
cConfirmation of the number of additional carbohydrate chains was made by examining the glycoprotein migration in SDS-gels after partial N-glycanase digestion as described in Example 7A.
dO-linked chain at Ser126 is present on 70% of human erythropoietin molecules.
eAnalogs having reduced O-glycosylation have a carbohydrate chain at Ser126 on less than 70% of the molecules.
fThr123 EPO has two O-linked chains on greater than 60% of the molecules. Thr125 EPO has two O-linked chains on about 40% of the molecules. Pro124Thr125 EPO has two O-linked chains on greater than 80% of the molecules.
gThese analogs have at least three O-linked chains and may have four or five. HCG alone is known to have four
O-linked chains.
N.T. NOT TESTED
TABLE 7
ACTIVITY OF HUMAN ERYTHROPOIETIN AND ANALOGS
HAVING CARBOHYDRATE SITE REARRANGEMENTS
In vi vo
N-Linked Activity
EPO Sequence Chains RIA or EIA
Human 3 0.69
Gln24 2 0.16
Gln38 2 0.16
Gln83 2 0.23
Gln24Ser87Asn88Thr90 (R1) 3 0.69
Gln38Ser87Asn88Thr90 (R2) 3 0.41
Gln83Ser87Asn88Thr90 (R3) 3 0.50
Ser87Asn88Thr90 3 to 4 1.48
Ratio of in vivo activity , to RIA or EIA was determined as described in footnote a of Table 6.
C. Preparation of Isoform Mixtures Derived from
Erythropoietin Analogs [Thr125] EPO (EPO O50)
The erythropoietin analog [Thr125] EPO was constructed as described in [Section A] Example 6. An 810 bp. erythropoietin cDNA fragment carrying the
[Thr125] mutation was isolated by cleaving the plasmid pEC containing the [Thr125] mutation with BstEII and BglII and ligating the fragment to pDECΔ, [a derivative of pDSα2] (described in Example 6).
Plasmid pDECΔ containing [Thr125]
erythropoietin cDNA was transfected into DHFR-deficient CHO cells. 770 ml of CHO cell conditioned medium was concentrated using a 10,000 dalton molecular weight cutoff membrane and diafiltered against 10 mM Tris-HCl, pH 8.6 to a final volume of 34 ml. A 17 ml. aliquot of the concentrate was loaded onto a Q-Sepharose fast flow column (5 ml bed volume) equilibrated in the same buffer and eluted in a linear gradient of 0-250 mM NaCI in 10 mM Tris-HCl, pH 8. 6. Aliquots of column fractions, either untreated or digested with N-glycanase, were analyzed by SDS-PAGE or IEF and pools (designated 2, 3 and 4) were made based upon the isoform and/or
carbohydrate composition of the fractions. Each pool was loaded onto a Vydac C4 column (214TPB 2030; 1 cm diameter; 1.8-2.5 ml bed volume; 0.34 ml/min) and washed with two column volumes of 20% ethanol in 10 mM Tris-HCl, pH 7.0. The columns were eluted with linear gradients of 20-94% ethanol, 10 mM Tris, pH 7.0. Pools were made, diluted into 10 mM Tris-HCl, pH 7.0, and loaded onto Q-Sepharose fast flow columns. Following a wash in 10 mM Tris-HCl, pH 7.0, the samples were eluted with 20 mM sodium citrate, 250 mM NaCI, pH 7.0. The purified [Thr125] pools were analyzed by IEF and are shown in Fig. 9. These pools were also analyzed for in vivo biological activity as described above (Cotes et al., supra) and the results shown in Table 8.
Ser87Asn88Thr90 EPO (EPO N14)
Prep,
The EPO N14 analog was purified using a three step procedure consisting of ion exchange
chromatography, reverse phase chromatography and gel filtration chromatography. During the ion exchange step, fractions were pooled to give a mixture of isoforms having high levels of sialic acid. Two additional pools were made during reverse phase chromatography containing the analog with or without aggregate. The details of the purification are set out below.
1. Conditional medium from CHO cells expressing the EPO N14 analog was harvested and
concentrated approximately 2.5-fold using a stirred cell with YM10 membrane (Amicon) and diafiltered against 10mM Tris pH 8.5.
2. The concentrated medium was loaded at ~12 A280/ml resin to Q-Sepharose FF (Pharmacia) column equilibrated in 10mM Tris pH 8.5 at a flow rate of 0.2 cm/min.
3. Following the load, the column was washed with 3 column volumes (CV) of 10mM Tris pH 8.5 and eluted with a gradient of 0-0.5M NaCl/10mM Tris pH 8.5 in 50CV. 1CV fractions were collected.
4. A sample of each fraction was run on an IEF gel pH 3-5. Based on the isoform distribution on IEF, a fraction pool containing largely isoforms 11-18 was made. EDTA was added to the pool to 1mM final concentration.
5. The isoform pool was loaded at ~5 A280/ml resin to a reverse phase C4 column (Vydac) equilibrated in 20% ethanol/10mM Tris pH 7.0 at a flow rate of 2.5 cm/min. The column was washed with 1CV of 20%
ethanol/10mM Tris pH 7.0 and eluted with a gradient of 20-94% ethanol/10mM Tris pH 7.0 in 30CV at a flow rate of lcm/min. 0.2CV fractions were collected.
6. A sample of each fraction was analyzed by non-reducing 12% SDS-PAGE. Two separate pools were made based on the presence of aggregate observed on SDS gels. Pool #1 contained EPO analog but no aggregate. Pool #2 contained both EPO analog and aggregate.
7. Pool #1 was concentrated approximately 65-fold and pool #2 approximately 250-fold using Centricon 10's (Amicon). Each pool was buffer exchanged into 20mM NaCitrate/100mM NaCI pH7.0.
8. Each pool was individually purified on HPLC BioSil SEC-250 (BioRad) column equilibrated in 20mM NaCitrate/100mM NaCI pH7.0. Pools were loaded at <6 A280/ml resin at a flow rate of 2.26 cm/min. Peaks corresponding to monomeric analogs were collected from each run.
9. The absorbance of each pool was measured and a portion of each pool was concentrated for analysis by SDS-PAGE and IEF gels. Pool #1 had a distribution of isoforms 15-17 and was used for pharmacokinetic and receptor binding studies. Prep. 2
EPO N14 analog was purified using a three step procedure consisting of ion exchange chromatography, reverse phase HPLC, and hydroxylapatite chromatography.
During the ion exchange step, the analog was divided into three pools containing different mixtures of isoforms. The details of the purification are set out below.
1. Supernatant from CHO cells expressing EPO N14 were harvested and concentrated approximately 10-fold using a Filtron Mini-Ultrasette tangential flow device and diafiltered against 10mM Tris pH 8.5.
2. The concentrated medium was loaded at ~10 A280/ml resin to Q-Sepharose FF (Pharmacia) column equilibrated in 10mM Tris pH 8.5 at a flow rate of 0.2 cm/min.
3. Following the load, the column was washed with three column volumes (CV) of 10mM Tris pH8.5 and eluted with a gradient of 0-0.5M NaCl/10mM Tris pH 8.5 in 50CV. 1 CV fractions were collected.
4. A sample of each fraction was run on an
IEF gel pH 3-5. Three separate fraction pools were prepared based on the isoform distribution as determined by IEF. A low isoform pool (isoform 4-12), a medium isoform pool (isoform 5-15), and a high isoform pool (isoform 6-18) were made.
5. Each isoform pool was individually
purified on a Vydac C4 reverse phase HPLC column. The column was developed with a gradient from 0.1% TFA/H2O to 0.1% TFA/75% acetonitrile at a rate of 1% increase in acetonitrile per minute.
6. The analog peak from each pool was
collected and diluted with 4 volumes 80mM Tris HCl/20mM Tris base, then concentrated and buffer exchanged into 10mM Tris pH 7.2 using solvent resistant Centricon 3's.
7. Each sample was diluted to 2 A280/ml in 10mM Tris pH 7.2 and an equal volume of 4M Guanidine HCI (GuHCl), 10mM CHAPS, 10mM Tris pH 7.2 was added for a final sample concentration of 1 A280/ml. Each sample was loaded onto a hydroxylapatite minicolumn equilibrated with 2M GuHCl, 5mM CHAPS, 10mM Tris pH 7.2. The column was washed with equilibration buffer and 1 CV fractions of flow through were collected.
8. The absorbance of fractions was measured and pools were made and buffer exchanged into 10mM Tris pH 7.2 using Centricon 10's. The absorbance of each pool was measured.
The final pools were analyzed by SDS-PAGE and IEF gels. The IEF gels are shown in Figure 12. The RIA and in vivo activity assays were performed as described in Example 7A. The in vivo activity of the final isoform pools is reported in Table 8. The high sialic acid isoform pool was used in experiments to determine the increase in the hematocrit of mice. TABLE 8
ACTIVITY OF ISOFORMS FROM ERYTHROPOIETIN ANALOGS
EPO Sequence Isoform Pool In vivo activity
u/mg. peptidea
Thr125 Isoforms 8-12 147,000b
Isoforms 9-15 215,000b
Isoforms 13-15 111,000b
Ser87Asn88Thr90 Isoforms 7-12 132,000 ± 17,000
Isoforms 10-14 233,000 ± 39,000 Isoforms 12-17 272,000 ± 14,000
amilligrams of erythropoietin peptide were calculated from A28O of the Ser87Asn88Thr90 EPO isoform pools using an extinction coefficient of 0.93 for a lmg/ml solution. bstandard deviations for Thr125 EPO isoform pools are not reported since activity assays were done either once or twice. Example 8: Biological Properties of EPO N14 Analog
The activities of isoform pools of EPO N14 analog, recombinant human erythropoietin (rHuEPO) and isolated rHuEPO isoform 14 were compared in i.v.
pharmacokinetic assays, receptor binding assays and hematocrit studies. EPO N14 isoform pools were prepared as described in Example 7C. rHuEPO was prepared
according to Lai et al. supra. rHuEPO isoform 14 was purified as follows: rHuEPO consisting of a mixture of isoforms 10, 11, 12, 13, 14 and 15 was loaded to a
Q-Sepharose Fast Flow column (Dimensions = 2.2 cm dia. X 3.4 cm ht.) equilibrated in 10 mM Tris pH 7.2. The loaded column was equilibrated to 2mM acetic acid / 6 M urea ("A" buffer) then a multiphasic gradient designed to optimize purification of isoforms 13 and 14 was run. The gradient was 0% to 7.3% 800 mM acetic acid / 6 M urea ("B" buffer) in 750 mL, 7.3% to 11.4% "B" buffer in 750 mL, 11.4% to 26.8% "B" buffer in 1250 mL, 26.8% to 62.4% "B" buffer in 1250 mL, then 62.4% to 100% "B" buffer in 700 mL. Fractions of 12.5 mL were collected, neutralized with ammonium hydroxide, then assayed by isoelectric focusing in polyacrylamide gels. Those fractions containing pure isoform 14 were pooled
together, concentrated with an Amicon stirred cell equipped with a YM-10 membrane then buffer exchange to water.
A. IV Pharmacokinetics
Two separate studies were performed to compare the pharmacokinetic parameters of EPO N14 analog
(isoforms 15-17) and isolated isoform 14 to rHuEPO.
In each study 1 μCi of either 125I-isolated isoform 14, 125I-EPO N14 analog, or 125I-recombinant human erythropoietin (Amersham) was injected
intravenously into a carotid cannula in male
Sprague-Dawley rats weighing between 310-378g. At various time points after administration, 0.3ml of blood was collected and serum was prepared by centrifugation. The 125I-EPO (either recombinant human, isolated isoform 14, or EPO N14 analog) concentration in 0.1ml of each serum sample was then determined following an overnight 4°C incubation with 90% ethanol. The ethanol-precipitated 125I-EPO in each serum sample was counted in a gamma counter and the resulting pharmacokinetic curves are shown in figure 13. The pharmacokinetic parameters were determined for each rat using PCNONLIN 4.0
nonlinear regression analysis (Statistical Consultants, 1992) and the results for each group were averaged. The results from the EPO N14 analog and the isolated isoform 14 pharmacokinetic studies are summarized in Table 9.
As seen in Table 9, there is a significant difference in the serum clearance of EPO N14 analog when compared to that calculated for recombinant human erythropoietin. The recombinant human erythropoietin displayed the fastest beta half-life of 3.10 hrs compared to 3.97 hrs for isolated isoform 14 and 4.36 hrs for EPO N14 analog. The recombinant human
erythropoietin group also had the fastest clearance half-life of 1.78 hrs compared to 2.40 hrs for isolated isoform 14 and 3.03 hrs for EPO N14 analog.
TABLE 9
SUMMARY OF THE PHARMACOKINETIC PARAMETERS OF rHuEPO, rHuEPO ISOFORM 14
AND EPO N14 (ISOFORMS 15-17) FOLLOWING I.V. ADMINISTRATION
Preparation Half-Life AUC VD Elimination Clearance Clearance
(Hours) (cpm-hr/ml) (mL/Kg) Rate Constant (ml/Kg-hr) Half-Life
Alpha Beta 0 - ∞ Hr. (Hr1) (Hours) rHuEPO Mean 0.332 3.10 308901 44.9 0.400 6.14 1.78
*n=6 S.D. 0.155 0.54 69187 8.3 0.069 1.36 0.27
Isoform 14 Mean 0.388 3.97 483329 36.5 0.288 3.80 2.40
n=4 S.D. 0.066 0.40 53638 2.2 0.020 0.12 0.15
EPO N14 Mean 0.422 4.36 692231 37.4 0.230 2.67 3.03
n=4 S.D. 0.165 0.36 40355 1.9 0.021 0.17 0.27 * The average of the recombinant human erythropoietin groups from the EPO N14
pharmacokinetic study (n=4 for the recombinant human erythropoietin group) and from the isolated isoform 14 pharmacokinetic study (n=2 for the recombinant human erythropoietin group).
B. Receptor Binding Assays
The interaction of EPO N14 analog (isoforms 15-17) with the erythropoietin receptor was studied by a cold displacement assay using human erythroleukemia 0CIM1 cells (Papayannopoulou et al. Blood 64 (supp.l), 116a (1984)). Increasing concentrations of unlabeled EPO N14 were incubated with OCIM1 cells along with a constant concentration of 125I-rHuEPO to determine the amount of EPO N14 required to compete with 125I rHuEPO for binding to the receptor. As a comparison,
increasing concentrations of unlabeled rHuEPO were also competed with a constant concentration of 125I-rHuEPO.
Unlabeled EPO N14 was diluted in assay buffer and added in amounts of 0.03ng, 0. lng, 0.3ng, 1.0ng, 3.0ng, 10.0ng, and 30.0ng (based on peptide mass).
0.5ng of 125I-recombinant human EPO were added to all assay tubes followed by the addition of approximately 0.5xl06 OCIM1 cells. The assay tubes were then
incubated at 37°C in a shaking water bath for 2 hours.
Following the incubation, the solutions were centrifuged through a dibutyl phthalate/bi-phthalate oil solution to separate unbound 125I-rHuEPO from 125I-rHuEPO bound to OCIM1 cells. Cell pellets were isolated and the amount of 125I-rHuEPO bound to the cells was determined by gamma counting. Counts specifically bound to the cells were calculated and the concentration of unlabeled protein required to compete 50% of the 125I-rHuEPO binding in the absence of competitor was determined by linear
regression analysis.
The results of three assays indicated that an average of 2.67 ± 0.73ng of unlabeled EPO N14 peptide was required to reduce 125I-rHuEPO binding to OCIM1 cells by 50%. In the same three assays, unlabeled rHuEPO required an average of 0.51 + 0.15 ng to compete with the binding of 0.5ng of 125I-rHuEPO. Based upon these results, a 5.25 fold excess of EPO N14 was required to compete the binding of 125I-rHuEP0 to the EPO receptor compared to unlabeled r-HuEPO (p<0.01).
An additional experiment was performed to make a direct comparison between EPO N14 analog, isolated isoform 14, and recombinant erythropoietin for binding to the erythropoietin receptor. The results of this experiment indicate that EPO N14 analog had a lower affinity for the receptor than isolated isoform 14. An approximate two-fold excess of EPO N14 analog was required to compete the binding of 125I-rHuEPO to the receptor compared to unlabeled isoform 14 (Figure 14). c Hematocrit Study
An in vivo study was performed to compare the ability of EPO N14 high and low isoform pools (from prep 2 described in Example 7C), isolated isoform 14, and recombinant human EPO to increase the hematocrit of treated mice. The isoform distribution of EPO N14 high and low isoform pools used in this study is shown in Figure 12.
CD1 mice (about 30g) were injected
intraperitoneally three times per week for a total of six weeks with one of the above preparations prepared in 0.25% mouse serum albumin, or with placebo (PBS with 0.25% mouse serum albumin). The dosage of
erythropoietin preparations was based on peptide mass, so that a 30g mouse received either 0.071μg of
peptide/dose for EPO N14 high pool, EPO N14 low pool, isoform 14, and r-HuEPO standard. An additional dosage group of 0.036μg of peptide/dose was included for EPO
N14 high pool and isoform 14. The hematocrits of all mice were determined at baseline and twice weekly thereafter by retroorbital bleeds. At the conclusion of the experiment, serum from all animals was collected and assayed for antibodies to the injected product. Data from animals judged to be negative for neutralizing antibodies was used for subsequent analysis.
As shown in figure 15, animals treated with the EPO N14 high isoform pool attained the highest group average hematocrits compared to the other preparations. Isoform 14, recombinant human EPO and the EPO N14 low isoform pool increased hematocrit to a lesser extent.
In order to compare the different
erythropoietin preparations in a more quantitative manner, the average initial rate of hematocrit rise (0-11 days), area under the curve (0-39 days), and the total hematocrit increase were calculated (Table 10). By any of these criteria, the EPO N14 high isoform pool appears to be more active on a peptide mass basis than any other preparation tested. Both the EPO N14 high isoform pool and isoform 14 were more active than recombinant human EPO. The EPO N14 low pool had the lowest activity when any of the analyses was used for comparison.
TABLE 10
EFFECT OF EPO N14 (HIGH AND LOW ISOFORM POOLS) , ISOFORM 14
AND rHuEPO ON THE HEMATOCRIT OF MICE
Preparation Dose (μg) Rate of Het Area Under Total Het
Rise1 Curve2 Increase3
(points/day) (Het points)
N14 High Pool 0.071 1.46 619 25.4
Isoform 14 0.071 1.19 546 22.6
N14 High Pool 0.036 0.98 428 19.1
Isoform 14 0.036 0.76 284 10.7 r-HuEPO 0.071 1.0 424 17.5
N14 Low Pool 0.071 0.5 212 9.3
1Initial rate of hematocrit rise was calculated from days 0-11 by linear regression.
2Area under the curve was calculated from- day 0-39 by trapezoidal summation.
3Total hematocrit increase was calculated as the group average hematocrit on day 39 minus group average hematocrit at baseline.
While the invention has been described in what is considered to be its preferred embodiments, it is not to be limited to the disclosed embodiments, but on the contrary, is intended to cover various modifications and equivalents included within the spirit and scope of the appended claims, which scope is to be accorded the broadest interpretation so as to encompass all such modifications and equivalents.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Elliott, Steven G.
Byrne, Thomas E.
(ii) TITLE OF INVENTION: Erythropoietin Analogs
(iii) NUMBER OF SEQUENCES: 26
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Amgen Inc., U.S. Patent Operations\rbw
(B) STREET: 1840 DeHavilland Drive
(C) CITY: Thousand Oaks
(D) STATE: California
(E) COUNTRY: U.S.A.
(F) ZIP: 91320-1789
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/108,016
(B) FILING DATE: 17-AUG-1993
(C) CLASSIFICATION:
(2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:
CGCCCACCAA ACCTCAGCTG TGACAGCCGA 30
(2) INFORMATION FOR SEQ ID NO: 2:
(i) SEQUENCE CHARACTERISTICS :
(A) LENGTH : 27 base pairs
(B) TYPE: nuclcic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:
ATCTGTACAA CCGAAGCCTG GAGAGGT 27
(2) INFORMATION FOR SEQ ID NO: 3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS : single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:
GGGCCTGGCC AACCTGTCGG AAG 23
(2) INFORMATION FOR SEQ ID NO: 4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 26 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:
TCCCCTCCAG ATAATGCCTC AGCTGC 26
(2) INFORMATION FOR SEQ ID NO: 5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5:
CAGATGCGAA CTCATCTGCT CCAC 24 (2) INFORMATION FOR SEQ ID NO: 6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6:
AGGCCTGCAG GAATGGGAGC AGATGACCAG GTG 33
(2) INFORMATION FOR SEQ ID NO: 7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7:
TCCAGATGCG ACCTCAGCTG CTC 23
(2) INFORMATION FOR SEQ ID NO: 8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:
CCTCCAGATC CGACCTCAGC TGC 23 (2) INFORMATION FOR SEQ ID NO: 9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 28 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
GGGCCTGGCC AACCTGACAG AAGCTGTC 28
(2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 30 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:
CAGGGCCTGT CCAACCTGAC AGAAGCTGTC 30
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
CAGATGCGAA CTCAACGGCT CCAC 24
(2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 35 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:
ATGCGAACTC AACGGCTCCA CTCACAACAA TCACT 35 (2) INFORMATION FOR SEQ ID NO: 13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13:
CCAGATCCAA ATTCATCTGC TCCACTC 27
(2) INFORMATION FOR SEQ ID NO: 14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 27 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:
CCAGATCCAA ATTCAACAGC TCCACTC 27
(2) INFORMATION FOR SEQ ID NO: 15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 24 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:
CCAGATGCGA CAACAGCTGC TCCA 24
(2) INFORMATION FOR SEQ ID NO: 16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16:
AGATCCGACC ACCGCTGCTC CAC 23
(2) INFORMATION FOR SEQ ID NO: 17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17:
TGCTCCACTC ACAACAATCA CTG 23
(2) INFORMATION FOR SEQ ID NO: 18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 184 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18:
TCGAGGAACT GAAAAACCAG AAAGTTAACT GGTAAGTTTA GTCTTTTTGT CTTTTATTTC 60
AGGTCCCGGA TCCGGTGGTG GTGCAAATCA AAGAACTGCT CCTCAGTGGA TGTTGCCTTT 120
ACTTCTAGGC CTGTACGGAA GTGTTACTTC TGCTCTAAAA GCTGCTGCAA CAAGCTGGTC 180
GACC 184
(2) INFORMATION FOR SEQ ID NO: 19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 107 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:
CCTGTAGGAC AGGGGACAGA TCCTCTTCCT CAAAGGCCCC TCCCCCCAGC CTTCCAAGTC 60 CATCCCGACT CCCGGGGCCC TCGGACACCC CGATCCTCCC ACAATGA 107
(2) INFORMATION FOR SEQ ID NO: 20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 108 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: double
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: CDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20:
GATCTCATTG TGGGAGGATC GGGGTGTCCG AGGGCCCCGG GAGTCGGGAT GGACTTGGAA 60 GGCTGGGGGG AGGGGCCGAG GAAGAGGATC TGTCCCCTGT CCTACAGG 108
(2) INFORMATION FOR SEQ ID NO: 21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21:
GGAGGCCGAG CAGATCACGA CGG 23 (2) INFORMATION FOR SEQ ID NO: 22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22:
CTTGAATGAG CAGATCACTG TCC 23
(2) INFORMATION FOR SEQ ID NO: 23:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 21 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23:
CTGTTGGTCC AGTCTTCCCA G 21
(2) INFORMATION FOR SEQ ID NO: 24:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:24:
CCTGTTGGTC CAGTCTTCCC AGC 23
(2) INFORMATION FOR SEQ ID NO: 25:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 28 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25:
Ser Ser Ser Ser Lys Ala Pro Pro Pro Ser Leu Pro Ser Pro Ser Arg 1 5 10 15
Leu Pro Gly Pro Ser Asp Thr Pro Ile Leu Pro Gln
20 25
(2) INFORMATION FOR SEQ ID NO: 26:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 193 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 26:
Met Gly Val His Glu Cys Pro Ala Trp Leu Trp Leu Leu Leu Ser Leu 1 5 10 15
Leu Ser Leu Pro Leu Gly Leu Pro Val Leu Gly Ala Pro Pro Arg Leu
20 25 30.
Ile Cys Asp Ser Arg Val Leu Glu Arg Tyr Leu Leu Glu Ala Lys Glu
35 40 45
Ala Glu Asn Ile Thr Thr Gly Cys Ala Glu His Cys Ser Leu Asn Glu 50 55 60
Asn Ile Thr Val Pro Asp Thr Lys Val Asn Phe Tyr Ala Trp Lys Arg 65 70 75 80
Met Glu Val Gly Gln Gln Ala Val Glu Val Trp Gln Gly Leu Ala Leu
85 90 95
Leu Ser Glu Ala Val Leu Arg Gly Gln Ala Leu Leu Val Asn Ser Ser
100 105 110
Gln Pro Trp Glu Pro Leu Gln Leu His Val Asp Lys Ala Val Ser Gly
115 120 125
Leu Arg Ser Leu Thr Thr Leu Leu Arg Ala Leu Gly Ala Gln Lys Glu 130 135 140
Ala Ile Ser Pro Pro Asp Ala Ala Ser Ala Ala Pro Leu Arg Thr Ile 145 150 155 160 Thr Ala Asp Thr Phe Arg Lys Leu Phe Arg Val Tyr Ser Asn Phe Leu 165 170 175
Arg Gly Lys Leu Lys Leu Tyr Thr Gly Glu Ala Cys Arg Thr Gly Asp
180 185 190
Arg

Claims

WHAT IS CLAIMED IS:
1. An analog of human erythropoietin comprising an amino acid sequence which includes at least one additional site for glycosylation.
2. The analog of Claim 1 wherein the glycosylation site is for an N-linked carbohydrate chain.
3. The analog of Claim 1 wherein the glycosylation site is for an O-linked carbohydrate chain.
4. The analog of Claim 1 having at least one additional carbohydrate chain attached thereto.
5. The analog of Claim 4 wherein the carbohydrate chain is an N-linked carbohydrate chain.
6. The analog of Claim 4 wherein the carbohydrate chain is an O-linked carbohydrate chain.
7. The analog of Claim 1 which is the product of expression of an exogenous DNA sequence.
8. The analog of Claim 2 wherein an aspargine residue is substituted at any of positions 30, 51, 57, 69, 88, 89, 136 or 138 of the amino acid sequence of human erythropoietin.
9. The analog of Claim 3 wherein a serine or threonine residue is substituted at position 125 of the amino acid sequence of human erythropoietin.
10 . An analog of human erythropoietin selected from the group consisting of :
Asn30Thr32 EPO;
Asn51Thr53 EPO;
Asn57Thr59 EPO;
Asn69EPO;
Asn69Thr71 EPO;
Ser68Asn69Thr71 EPO;
Val87Asn88 Thr90 EPO;
Ser87Asn88Thr9° EPO;
Ser87Asn88Gly89Thr90 EPO;
Ser87Asn88Thr90Thr92 EPO;
Ser87Asn88Thr90Ala162 EPO;
Asn69Thr71Ser87Asn88Thr90 EPO;
Asn30Thr32Val87Asn88Thr90 EPO;
Asn89Ile90Thr91 EPO;
Ser87Asn89 Ile90Thr91 EPO;
Asn136Thr138 EPO;
Asn138Thr140 EPO;
Thr125 EPO; and
Pro124Thr125 EPO .
11. An analog comprising an addition of one or more amino acids to the carboxy terminal end of
erythropoietin wherein the addition includes at least one glycosylation site.
12. The analog of Claim 11 wherein the addition comprises a peptide fragment derived from the carboxy terminal end of human chorionic gonadotropin.
13. The analog of Claim 12 selected from the group consisting of:
(a) human erythropoietin having the amino acid sequence Ser-Ser-Ser-Ser-Lys-Ala-Pro-Pro-Pro-Ser-Leu- Pro-Ser-Pro-Ser-Arg-Leu-Pro-Gly-Pro-Ser-Asp-Thr-Pro-Ile-Leu-Pro-Gln extending from the carboxy terminus;
(b) the analog in (a) further comprising Ser87Asn88Thr90 EPO; and
(c) the analog in (a) further comprising
Asn30Thr32Val87Asn88Thr90 EPO.
14. An analog of human erythropoietin comprising an amino acid sequence which includes a rearrangement of at least one site for glycosylation.
15. The analog of Claim 14 wherein the
rearrangement comprises a deletion of any of the N-linked carbohydrate sites in human erythropoietin and an addition of an N-linked carbohydrate site at position 88 of the amino acid sequence of human erythropoietin.
16. The analog of Claim 15 selected from the group consisting of:
Gln24Ser87Asn88Thr90 EPO;
Gln38Ser87Asn88Thr90 EPO; and
Gln83Ser87Asn88Thr90 EPO.
17. A DNA sequence encoding an analog of human erythropoietin which includes at least one additional site for glycosylation.
18. A DNA sequence encoding an analog of human erythropoietin according to Claim 10.
19. A DNA sequence encoding an analog of human erythropoietin having a rearrangement of at least one site for glycosylation.
20. A DNA sequence encoding an analog of human erythropoietin according to Claim 16.
21. A DNA sequence encoding an analog of human erythropoietin according to Claim 13.
22. A eucaryotic host cell transfected with a
DNA sequence according to either of Claims 17 or 19 in a manner allowing the host cell to express an analog of human erythropoietin.
23. A composition comprising a therapeutically effective amount of an erythropoietin analog according to any of Claims 1, 10, 11 or 14 together with a
pharmaceutically acceptable diluent, adjuvant or carrier.
PCT/US1994/009257 1993-08-17 1994-08-16 Erythropoietin analogs WO1995005465A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
CA002147124A CA2147124C (en) 1993-08-17 1994-08-16 Erythropoietin analogs
KR1019950701453A KR100328769B1 (en) 1993-08-17 1994-08-16 Erythropoietin analog
UA95058432A UA49793C2 (en) 1993-08-17 1994-08-16 Human erythropoietin analogs (variants), dna sequences encoding erythropoietin analog (variants), eukaryotic host cells transfected with plasmid dna (variants), pharmaceutical formulation (variants)
HU9501435A HU220334B (en) 1993-08-17 1994-08-16 Erythropoietin analogs
JP7507153A JP2938572B2 (en) 1993-08-17 1994-08-16 Erythropoietin analog
SK502-95A SK282003B6 (en) 1993-08-17 1994-08-16 Human erythropoietin analogs, dna sequence, eucaryotic host cell and pharmaceutical preparation
NZ273134A NZ273134A (en) 1993-08-17 1994-08-16 Erythropoietin analogs, their production and compositions thereof
AU76327/94A AU677097B2 (en) 1993-08-17 1994-08-16 Erythropoietin analogs
NO19951445A NO323104B1 (en) 1993-08-17 1995-04-12 Analogs of human erythropoietin, DNA sequences encoding them, eukaryotic host cell transfected with the DNA sequences, preparations comprising the analogs, use of the analogs to increase hematocrit, and methods for preparing the analogs.
FI951792A FI117136B (en) 1993-08-17 1995-04-13 A process for the preparation of therapeutically useful erythropoietin analogs
KR1019957001453A KR960701994A (en) 1993-08-17 1995-04-15 ERYTHROPOIETIN ANALOGS
LVP-95-99A LV10972B (en) 1993-08-17 1995-04-17 Erythropoietin analogs
NO2007008C NO2007008I2 (en) 1993-08-17 2007-06-29 Analogs of human erythropoietin, DNA sequences encoding them, eukaryotic host cell transfected with the DNA sequences, preparations comprising the analogs, use of the analogues to increase hematocrit, and method of preparing the analogs.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10801693A 1993-08-17 1993-08-17
US08/108,016 1993-08-17

Publications (1)

Publication Number Publication Date
WO1995005465A1 true WO1995005465A1 (en) 1995-02-23

Family

ID=22319789

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/009257 WO1995005465A1 (en) 1993-08-17 1994-08-16 Erythropoietin analogs

Country Status (27)

Country Link
EP (1) EP0640619B2 (en)
JP (3) JP2938572B2 (en)
KR (2) KR100328769B1 (en)
CN (1) CN1057534C (en)
AT (1) ATE155796T1 (en)
AU (1) AU677097B2 (en)
CA (1) CA2147124C (en)
CZ (3) CZ291229B6 (en)
DE (2) DE10199059I2 (en)
DK (1) DK0640619T4 (en)
ES (1) ES2105442T5 (en)
FI (1) FI117136B (en)
GR (1) GR3024815T3 (en)
HK (1) HK1001589A1 (en)
HU (1) HU220334B (en)
IL (4) IL110669A (en)
LU (1) LU90850I2 (en)
LV (1) LV10972B (en)
NL (1) NL300075I2 (en)
NO (2) NO323104B1 (en)
NZ (1) NZ273134A (en)
RU (1) RU2159814C2 (en)
SI (1) SI0640619T2 (en)
SK (1) SK282003B6 (en)
UA (1) UA49793C2 (en)
WO (1) WO1995005465A1 (en)
ZA (1) ZA946122B (en)

Cited By (156)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997013783A1 (en) * 1995-10-09 1997-04-17 Pharmacia & Upjohn Company An acceptor polypeptide for an n-acetylgalactosaminyltransferase
US5696250A (en) * 1995-02-15 1997-12-09 Amgen Inc. DNA encoding megakaryocyte growth and development factor analogs
US5989538A (en) * 1995-02-15 1999-11-23 Amgen Inc. Mpl ligand analogs
US6048971A (en) * 1992-07-28 2000-04-11 Beth Israel Deaconess Medical Center Recombinant human erythropoietin mutants
WO2000024893A2 (en) * 1998-10-23 2000-05-04 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
WO2000047741A1 (en) * 1999-02-12 2000-08-17 Amgen Inc. Glycosylated leptin compositions and related methods
WO2000068376A1 (en) * 1999-05-07 2000-11-16 Genentech, Inc. Novel chimpanzee erythropoietin (chepo) polypeptides and nucleic acids encoding the same
US6504007B1 (en) 1996-03-14 2003-01-07 Genentech, Inc. GDNF receptor
US6531121B2 (en) 2000-12-29 2003-03-11 The Kenneth S. Warren Institute, Inc. Protection and enhancement of erythropoietin-responsive cells, tissues and organs
US6548653B1 (en) 1998-06-15 2003-04-15 Genzyme Transgenics Corporation Erythropoietin analog-human serum albumin fusion
US6555343B1 (en) 1999-05-07 2003-04-29 Genentech Inc. Chimpanzee erythropoietin (CHEPO) polypeptides and nucleic acids encoding the same
US6583272B1 (en) * 1999-07-02 2003-06-24 Hoffmann-La Roche Inc. Erythropoietin conjugates
US6586398B1 (en) * 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
KR100400637B1 (en) * 1996-02-06 2004-03-10 씨제이 주식회사 Biologically useful peptide
US6831060B2 (en) 1999-05-07 2004-12-14 Genentech, Inc. Chimpanzee erythropoietin (CHEPO) polypeptides and nucleic acids encoding the same
DE10235248B4 (en) * 2001-11-29 2004-12-23 Cheil Jedang Corp. Fusion protein with enhanced in vivo activity of erythropoietin
US7041794B2 (en) * 2001-04-04 2006-05-09 Genodyssee Polynucleotides and polypeptides of the erythropoietin gene
US7091326B2 (en) 2001-12-03 2006-08-15 Cheil Jedang Corporation Fusion protein having enhanced in vivo erythropoietin activity
US7132280B2 (en) 1999-04-15 2006-11-07 Crucell Holland, B.V. Recombinant protein production in a human cell
WO2006120030A1 (en) 2005-05-13 2006-11-16 Charite Universitätsmedizin - Berlin Erythropoietin variants
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
WO2007108505A1 (en) 2006-03-22 2007-09-27 Chugai Seiyaku Kabushiki Kaisha Erythropoietin solution preparation
US7291484B2 (en) 2003-05-09 2007-11-06 Crucell Holland B.V. Processes for culturing E1-immortalized cells to increase product yields
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
US7304150B1 (en) 1998-10-23 2007-12-04 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
US7309687B1 (en) 1999-04-13 2007-12-18 The Kenneth S. Warren Institute, Inc. Methods for treatment and prevention of neuromuscular and muscular conditions by peripherally administered erythropoietin
WO2008023725A1 (en) 2006-08-22 2008-02-28 Chugai Seiyaku Kabushiki Kaisha Preventive and/or therapeutic agent for peripheral neuropathy
US7345019B1 (en) 1999-04-13 2008-03-18 The Kenneth S. Warren Institute, Inc. Modulation of excitable tissue function by peripherally administered erythropoietin
WO2008111503A1 (en) 2007-03-09 2008-09-18 National University Corporation Obihiro University Of Agriculture And Veterinary Medicine Protective agent for transplanted organ
US7504248B2 (en) 2001-12-07 2009-03-17 Crucell Holland B.V. Production of viruses viral isolates and vaccines
US7514072B1 (en) 1998-12-14 2009-04-07 Hannelore Ehrenreich Method for the treatment of cerebral ischaemia and use of erythropoietin or erythropoietin derivatives for the treatment of cerebral ischaemia
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
WO2009094551A1 (en) 2008-01-25 2009-07-30 Amgen Inc. Ferroportin antibodies and methods of use
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
EP2156843A2 (en) 2004-01-23 2010-02-24 EPOPLUS GmbH &amp; Co. KG Use of low-dose erythropoietin for the treatment of acute or chronic renal insufficiency and for the treatment of wounds
WO2010056981A2 (en) 2008-11-13 2010-05-20 Massachusetts General Hospital Methods and compositions for regulating iron homeostasis by modulation bmp-6
EP2191838A1 (en) 2002-07-26 2010-06-02 EPOPLUS GmbH &amp; Co. KG Erythropoetin for the stimulation of endothelial precursor cells
US7847079B2 (en) 2001-12-21 2010-12-07 Human Genome Sciences, Inc. Albumin fusion proteins
US7851438B2 (en) 2006-05-19 2010-12-14 GlycoFi, Incorporated Erythropoietin compositions
WO2011011674A2 (en) 2009-07-24 2011-01-27 Dr. Reddy's Laboratories Ltd. Production of erythropoiesis stimulating protein using metal ions
EP2281828A2 (en) 2000-12-29 2011-02-09 The Kenneth S. Warren Institute, Inc. Compositions comprising modified erythropoietin
US7888101B2 (en) 2005-12-08 2011-02-15 Amgen Inc. Host cells comprising alpha 1,2 mannosidase and culture methods thereof
WO2011050333A1 (en) 2009-10-23 2011-04-28 Amgen Inc. Vial adapter and system
EP2319530A1 (en) 2006-01-27 2011-05-11 Hannelore Ehrenreich Method for treating and/or preventing multiple sclerosis, and use of erythropoietin in the production of a drug for intermittent treatment and/or intermittent prevention of multiple sclerosis
US7972810B2 (en) 2005-12-08 2011-07-05 Amgen Inc. Production of glycoproteins using manganese
WO2011156373A1 (en) 2010-06-07 2011-12-15 Amgen Inc. Drug delivery device
US8114839B2 (en) * 2002-09-09 2012-02-14 Hanall Biopharma Co., Ltd. Protease resistant modified erythropoietin polypeptides
US8236561B2 (en) 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
EP2498096A1 (en) 2006-07-21 2012-09-12 Amgen Inc. Method of detecting and/or measuring hepcidin in a sample
WO2012135315A1 (en) 2011-03-31 2012-10-04 Amgen Inc. Vial adapter and system
CN102788720A (en) * 2012-06-05 2012-11-21 西北大学 Membrane assisted separation of glycoprotein all N-linked carbohydrate chain and identification method thereof
WO2013055873A1 (en) 2011-10-14 2013-04-18 Amgen Inc. Injector and method of assembly
EP2620448A1 (en) 2008-05-01 2013-07-31 Amgen Inc. Anti-hepcidin antibodies and methods of use
US8535923B2 (en) 2009-04-27 2013-09-17 Cytonet Gmbh & Co. Kg Encapsulated liver cell composition
US8633153B2 (en) 2002-04-04 2014-01-21 Amgen Inc. Transthyretin variants
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8716239B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
WO2014081780A1 (en) 2012-11-21 2014-05-30 Amgen Inc. Drug delivery device
WO2014144096A1 (en) 2013-03-15 2014-09-18 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
WO2014149357A1 (en) 2013-03-22 2014-09-25 Amgen Inc. Injector and method of assembly
US8853161B2 (en) 2003-04-09 2014-10-07 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8895303B2 (en) 2006-11-13 2014-11-25 Charite-Universitatsmedizin Berlin Method of cell culture and method of treatment comprising a vEPO protein variant
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
WO2015061386A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Injector and method of assembly
WO2015061389A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Drug delivery system with temperature-sensitive control
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
WO2015080509A1 (en) 2013-11-29 2015-06-04 씨제이헬스케어 주식회사 Method for purifying darbepoetin alfa
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
US9180165B2 (en) 2007-07-19 2015-11-10 Hannelore Ehrenreich Use of EPO receptor activation or stimulation for the improvement of the EDSS score in patients with multiple sclerosis
WO2015171777A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
WO2015187799A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Systems and methods for remotely processing data collected by a drug delivery device
WO2016049036A1 (en) 2014-09-22 2016-03-31 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof
WO2016061220A2 (en) 2014-10-14 2016-04-21 Amgen Inc. Drug injection device with visual and audio indicators
US9320797B2 (en) 2007-09-27 2016-04-26 Amgen Inc. Pharmaceutical formulations
WO2016100781A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with proximity sensor
WO2016100055A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with live button or user interface field
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
US9657098B2 (en) 2013-03-15 2017-05-23 Intrinsic Lifesciences, Llc Anti-hepcidin antibodies and uses thereof
WO2017100501A1 (en) 2015-12-09 2017-06-15 Amgen Inc. Auto-injector with signaling cap
WO2017120178A1 (en) 2016-01-06 2017-07-13 Amgen Inc. Auto-injector with signaling electronics
WO2017160799A1 (en) 2016-03-15 2017-09-21 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
WO2017189089A1 (en) 2016-04-29 2017-11-02 Amgen Inc. Drug delivery device with messaging label
WO2017192287A1 (en) 2016-05-02 2017-11-09 Amgen Inc. Syringe adapter and guide for filling an on-body injector
WO2017197222A1 (en) 2016-05-13 2017-11-16 Amgen Inc. Vial sleeve assembly
WO2017200989A1 (en) 2016-05-16 2017-11-23 Amgen Inc. Data encryption in medical devices with limited computational capability
WO2017209899A1 (en) 2016-06-03 2017-12-07 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
WO2018004842A1 (en) 2016-07-01 2018-01-04 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
WO2018081234A1 (en) 2016-10-25 2018-05-03 Amgen Inc. On-body injector
US9988427B2 (en) 2005-05-13 2018-06-05 Charite Universitaetsmedizen-Berlin Erythropoietin variants
WO2018136398A1 (en) 2017-01-17 2018-07-26 Amgen Inc. Injection devices and related methods of use and assembly
WO2018152073A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Insertion mechanism for drug delivery device
WO2018151890A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Drug delivery device with sterile fluid flowpath and related method of assembly
WO2018165499A1 (en) 2017-03-09 2018-09-13 Amgen Inc. Insertion mechanism for drug delivery device
WO2018164829A1 (en) 2017-03-07 2018-09-13 Amgen Inc. Needle insertion by overpressure
WO2018165143A1 (en) 2017-03-06 2018-09-13 Amgen Inc. Drug delivery device with activation prevention feature
WO2018172219A1 (en) 2017-03-20 2018-09-27 F. Hoffmann-La Roche Ag Method for in vitro glycoengineering of an erythropoiesis stimulating protein
EP3381445A2 (en) 2007-11-15 2018-10-03 Amgen Inc. Aqueous formulation of antibody stablised by antioxidants for parenteral administration
WO2018183039A1 (en) 2017-03-28 2018-10-04 Amgen Inc. Plunger rod and syringe assembly system and method
WO2018226515A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
WO2018226565A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Torque driven drug delivery device
WO2018237225A1 (en) 2017-06-23 2018-12-27 Amgen Inc. Electronic drug delivery device comprising a cap activated by a switch assembly
WO2018236619A1 (en) 2017-06-22 2018-12-27 Amgen Inc. Device activation impact/shock reduction
WO2019014014A1 (en) 2017-07-14 2019-01-17 Amgen Inc. Needle insertion-retraction system having dual torsion spring system
WO2019018169A1 (en) 2017-07-21 2019-01-24 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
WO2019022951A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with gear module and related method of assembly
WO2019022950A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with container access system and related method of assembly
WO2019032482A2 (en) 2017-08-09 2019-02-14 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
WO2019036181A1 (en) 2017-08-18 2019-02-21 Amgen Inc. Wearable injector with sterile adhesive patch
WO2019040548A1 (en) 2017-08-22 2019-02-28 Amgen Inc. Needle insertion mechanism for drug delivery device
WO2019070552A1 (en) 2017-10-06 2019-04-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
WO2019070472A1 (en) 2017-10-04 2019-04-11 Amgen Inc. Flow adapter for drug delivery device
WO2019074579A1 (en) 2017-10-09 2019-04-18 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
WO2019089178A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Drug delivery device with placement and flow sensing
WO2019090086A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Systems and approaches for sterilizing a drug delivery device
WO2019090303A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Fill-finish assemblies and related methods
WO2019094138A1 (en) 2017-11-10 2019-05-16 Amgen Inc. Plungers for drug delivery devices
WO2019099322A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Autoinjector with stall and end point detection
WO2019099324A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Door latch mechanism for drug delivery device
US10428129B2 (en) 2012-12-18 2019-10-01 Ubi Pharma Inc. Recombinant protein
EP3556411A1 (en) 2015-02-17 2019-10-23 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
WO2019231582A1 (en) 2018-05-30 2019-12-05 Amgen Inc. Thermal spring release mechanism for a drug delivery device
WO2019231618A1 (en) 2018-06-01 2019-12-05 Amgen Inc. Modular fluid path assemblies for drug delivery devices
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
WO2020023444A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020023451A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
WO2020028009A1 (en) 2018-07-31 2020-02-06 Amgen Inc. Fluid path assembly for a drug delivery device
WO2020068623A1 (en) 2018-09-24 2020-04-02 Amgen Inc. Interventional dosing systems and methods
WO2020065576A1 (en) 2018-09-27 2020-04-02 Universidad Nacional Del Litoral Modified human erythropoietin
WO2020068476A1 (en) 2018-09-28 2020-04-02 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
WO2020072846A1 (en) 2018-10-05 2020-04-09 Amgen Inc. Drug delivery device having dose indicator
WO2020072577A1 (en) 2018-10-02 2020-04-09 Amgen Inc. Injection systems for drug delivery with internal force transmission
WO2020081479A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Drug delivery device having damping mechanism
WO2020081480A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Platform assembly process for drug delivery device
WO2020091956A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020092056A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial needle retraction
WO2020219482A1 (en) 2019-04-24 2020-10-29 Amgen Inc. Syringe sterilization verification assemblies and methods
WO2021041067A2 (en) 2019-08-23 2021-03-04 Amgen Inc. Drug delivery device with configurable needle shield engagement components and related methods
EP3981450A1 (en) 2015-02-27 2022-04-13 Amgen, Inc Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
EP4074355A1 (en) 2011-04-20 2022-10-19 Amgen Inc. Autoinjector apparatus
WO2022246055A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container
WO2023209074A1 (en) 2022-04-28 2023-11-02 Institut National de la Santé et de la Recherche Médicale Methods of restoring erythropoiesis in patients suffering from a sf3b1 mutant myelodysplastic syndrome by correcting coasy mis-splicing

Families Citing this family (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7217689B1 (en) 1989-10-13 2007-05-15 Amgen Inc. Glycosylation analogs of erythropoietin
US5952226A (en) * 1996-11-05 1999-09-14 Modex Therapeutiques Hypoxia responsive EPO producing cells
PT902085E (en) * 1997-09-01 2004-02-27 Aventis Pharma Gmbh RECOMBINANT HUMAN ERYTHROPOIETIN WITH ADVANCED GLYCOSILACA PROFILE
ATE267215T1 (en) 1997-12-08 2004-06-15 Lexigen Pharm Corp HETERODIMARY FUSION PROTEINS FOR USE FOR TARGETED IMMUNTHERAPY AND GENERAL IMMUNE EXCITATION
JO2291B1 (en) * 1999-07-02 2005-09-12 اف . هوفمان لاروش ايه جي Erythopintin derivatives
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
WO2001036489A2 (en) * 1999-11-12 2001-05-25 Merck Patent Gmbh Erythropoietin forms with improved properties
CA2399832C (en) 2000-02-11 2011-09-20 Stephen D. Gillies Enhancing the circulating half-life of antibody-based fusion proteins
AU2001255516B2 (en) * 2000-04-21 2007-01-18 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
NZ522030A (en) * 2000-05-15 2004-11-26 F Erythropoietin composition with a multiple charged inorganic anion i.e. a sulfate a citrate or a phosphate to stabilize the composition
US7259146B2 (en) 2000-05-26 2007-08-21 Ortho-Mcneil Pharmaceutical, Inc. Neuroprotective peptides
JP5490972B2 (en) 2000-08-04 2014-05-14 中外製薬株式会社 Protein injection formulation
DK1314437T3 (en) 2000-08-11 2014-07-14 Chugai Pharmaceutical Co Ltd Stabilized antibody-containing preparations
DE60117919T2 (en) * 2000-12-11 2006-12-14 Cheil Jedang Corp. FUSION PROTEIN WITH IMPROVED IN VIVO ERYTHROPOIETING EFFECT
KR101229995B1 (en) 2000-12-11 2013-02-06 씨제이 주식회사 Fusion protein having the enhanced in vivo activity of erythropoietin
KR100645843B1 (en) 2000-12-20 2006-11-14 에프. 호프만-라 로슈 아게 Erythropoietin conjugates
US7767643B2 (en) 2000-12-29 2010-08-03 The Kenneth S. Warren Institute, Inc. Protection, restoration, and enhancement of erythropoietin-responsive cells, tissues and organs
US20030072737A1 (en) 2000-12-29 2003-04-17 Michael Brines Tissue protective cytokines for the protection, restoration, and enhancement of responsive cells, tissues and organs
DE60219961T8 (en) 2001-02-02 2008-04-17 Ortho-Mcneil Pharmaceutical, Inc. TREATMENT OF NEUROLOGICAL FUNCTIONAL DISORDERS WITH FRUCTOPYRANOSESULFAMATE AND ERYTHROPOETIN
CA2440221C (en) 2001-03-07 2013-02-05 Merck Patent Gesellschaft Mit Beschraenkter Haftung Expression technology for proteins containing a hybrid isotype antibody moiety
WO2002072615A1 (en) 2001-03-09 2002-09-19 Chugai Seiyaku Kabushiki Kaisha Method of purifying protein
DE10112825A1 (en) 2001-03-16 2002-10-02 Fresenius Kabi De Gmbh HESylation of active ingredients in aqueous solution
WO2002079415A2 (en) 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
EP2975382B1 (en) 2001-04-17 2017-12-27 Chugai Seiyaku Kabushiki Kaisha Method of quantifying surfactant
BR0209177A (en) 2001-05-03 2004-10-05 Merck Patent Gmbh Recombinant tumor specific antibody and use
US6930086B2 (en) 2001-09-25 2005-08-16 Hoffmann-La Roche Inc. Diglycosylated erythropoietin
US6818613B2 (en) 2001-11-07 2004-11-16 Ortho-Mcneil Pharmaceutical, Inc. Aqueous sustained-release formulations of proteins
JP2005530678A (en) 2001-11-28 2005-10-13 オーソーマクニール ファーマシューティカル, インコーポレイテッド Erythropoietin administration therapy for anemia treatment
ES2381025T3 (en) 2001-12-04 2012-05-22 Merck Patent Gmbh Immunocytokines with modulated selectivity
DE10209822A1 (en) 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of low molecular weight substances to a modified polysaccharide
DE10209821A1 (en) 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of proteins to a modified polysaccharide
US7129267B2 (en) 2002-03-11 2006-10-31 Janssen Pharmaceutica N.V. Methods for SHP1 mediated neuroprotection
BR0312395A (en) * 2002-07-01 2007-06-19 Kenneth S Warren Inst Inc recombinant mutein protective tissue cytokine, mammalian cell responsive recombinant tissue protective cytokine, isolated nucleic acid molecule, vector, expression vector, genetically engineered cell, cell, pharmaceutical composition, method for protecting, maintaining or enhancing the viability of a cell, tissue or organ isolated from a mammalian body, use of a recombinant tissue protective cytokine, method for facilitating the transcytosis of a molecule through an endothelial cell barrier in a mammal, and composition for transporting a molecule through of transcytosis through an endothelial cell barrier
AU2003246486A1 (en) * 2002-07-19 2004-02-09 Cangene Corporation Pegylated erythropoietic compounds
WO2004019966A1 (en) 2002-08-27 2004-03-11 Chugai Seiyaku Kabushiki Kaisha Method of stabilizing protein solution preparation
US7459435B2 (en) 2002-08-29 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
EP1681303B1 (en) 2002-09-11 2013-09-04 Fresenius Kabi Deutschland GmbH HASylated polypeptides, especially HASylated erythropoietin
US8420789B2 (en) 2002-09-11 2013-04-16 Chugai Seiyaku Kabushiki Kaisha Method for removing DNA contaminants from a protein-containing sample
WO2004024776A1 (en) 2002-09-11 2004-03-25 Fresenius Kabi Deutschland Gmbh Method of producing hydroxyalkyl starch derivatives
DE20321793U1 (en) 2002-09-11 2010-06-02 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives
AU2003273413A1 (en) 2002-10-08 2004-05-04 Fresenius Kabi Deutschland Gmbh Pharmaceutically active oligosaccharide conjugates
NZ539472A (en) * 2002-10-17 2008-05-30 Pharming Intellectual Pty Bv Protein modification
US7459436B2 (en) 2002-11-22 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
ES2346205T3 (en) 2002-12-17 2010-10-13 Merck Patent Gmbh HUMANIZED ANTIBODY (H14.18) OF ANTIBODY 14.18 OF MOUSE THAT LINKS TO GD2 AND ITS FUSION WITH IL-2.
WO2005014655A2 (en) 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
ZA200603396B (en) 2003-09-29 2007-11-28 Warren Pharmaceuticals Inc Tissue protective cytokines for the treatment and prevention of sepsis and the formation of adhesions
US7910547B2 (en) 2003-12-30 2011-03-22 Augustinus Bader Tissue regeneration method
TWI417303B (en) 2004-03-11 2013-12-01 Fresenius Kabi De Gmbh Conjugates of hydroxyalkyl starch and a protein, prepared by reductive amination
WO2006010143A2 (en) 2004-07-13 2006-01-26 Neose Technologies, Inc. Branched peg remodeling and glycosylation of glucagon-like peptide-1 [glp-1]
US7597884B2 (en) 2004-08-09 2009-10-06 Alios Biopharma, Inc. Hyperglycosylated polypeptide variants and methods of use
CN100432107C (en) * 2005-10-25 2008-11-12 北京大学 Protein for promoting erythrocyte growth factor activity and its use
CN101062407A (en) 2006-04-29 2007-10-31 中国科学院上海生命科学研究院 Function of erythropoietin in the preventing and treating of retinal injury
CN101337988B (en) * 2006-10-31 2013-01-09 沈阳三生制药有限责任公司 New erythrocyte-stimulating factor analogues
JP2010510794A (en) 2006-11-28 2010-04-08 ハナル ファーマシューティカル カンパニー リミテッド Modified erythropoietin polypeptide and therapeutic use thereof
JP2010534194A (en) * 2007-06-15 2010-11-04 チューリッヒ大学 Novel treatment for nervous system disorders
CN103113464B (en) * 2007-07-02 2014-08-20 沈阳三生制药有限责任公司 Natural human erythropoietin analogue
CN103073631A (en) * 2007-07-02 2013-05-01 沈阳三生制药有限责任公司 Hemopoietin analogue
EP2070950A1 (en) 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Hydroxyalkyl starch derivatives and process for their preparation
NZ620606A (en) 2008-02-08 2015-08-28 Ambrx Inc Modified leptin polypeptides and their uses
EP2095829A1 (en) 2008-02-27 2009-09-02 LEK Pharmaceuticals D.D. Selenium containing modifying agents and conjugates
JP5665733B2 (en) 2008-04-21 2015-02-04 ノボ ノルディスク ヘルス ケア アーゲー Highly glycosylated human coagulation factor IX
US20120172299A1 (en) 2008-09-23 2012-07-05 F. Hoffmann-La Roche Ag Purification of erythropoietin
CN107022020A (en) 2008-09-26 2017-08-08 Ambrx公司 The animal erythropoietin polypeptides and its purposes of modification
US9150894B2 (en) * 2009-02-19 2015-10-06 Xellia Pharmaceuticals Aps Process for purifying lipopeptides
WO2011024025A1 (en) * 2009-08-28 2011-03-03 Avesthagen Limited An erythropoietin analogue and a method thereof
US20120115637A1 (en) 2010-06-30 2012-05-10 Nike, Inc. Golf Balls Including A Crosslinked Thermoplastic Polyurethane Cover Layer Having Improved Scuff Resistance
US8193296B2 (en) 2010-06-30 2012-06-05 Nike, Inc. Golf balls including crosslinked thermoplastic polyurethane
RU2451071C1 (en) * 2010-12-10 2012-05-20 Российская Федерация, от имени которой выступает Министерство образования и науки Российской Федерации (Минобрнауки России) Mus musculus cultivated hybrid cell strain producer of monoclonal antibodies specific to human recombinant erythropoietin (versions)
US8979676B2 (en) 2011-08-23 2015-03-17 Nike, Inc. Multi-core golf ball having increased initial velocity at high swing speeds relative to low swing speeds
US9089739B2 (en) 2011-08-23 2015-07-28 Nike, Inc. Multi-core golf ball having increased initial velocity
KR101443257B1 (en) * 2011-10-18 2014-09-19 주식회사 종근당 Methods for Purifying Erythropoietin Analogs Having Lower Isoelectric Point
WO2016044676A2 (en) 2014-09-18 2016-03-24 AskGene Pharma, Inc. Novel feline erythropoietin receptor agonists
EP3428284B1 (en) * 2016-03-09 2022-05-11 JCR Pharmaceuticals CO., LTD. Method for producing mutant human erythropoietin
GB2550418A (en) 2016-05-20 2017-11-22 Laing Peter An improved vaccine against flaviviruses avoiding elicitation or stimulation of infection-enhancing antibodies
JP6258536B1 (en) * 2017-03-03 2018-01-10 協和発酵キリン株式会社 Method for producing darbepoetin composition and method for culturing darbepoetin-producing cells
US11667686B2 (en) 2017-06-06 2023-06-06 Kindred Biosciences, Inc. Erythropoietin and analogs for veterinary use
WO2022159414A1 (en) 2021-01-22 2022-07-28 University Of Rochester Erythropoietin for gastroinfestinal dysfunction

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0267678A1 (en) * 1986-09-15 1988-05-18 Genzyme Corporation Recombinant human erythropoietin
EP0428267A2 (en) * 1989-10-13 1991-05-22 Amgen Inc. Erythropoietin isoforms

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ210501A (en) * 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0267678A1 (en) * 1986-09-15 1988-05-18 Genzyme Corporation Recombinant human erythropoietin
EP0428267A2 (en) * 1989-10-13 1991-05-22 Amgen Inc. Erythropoietin isoforms

Cited By (254)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6048971A (en) * 1992-07-28 2000-04-11 Beth Israel Deaconess Medical Center Recombinant human erythropoietin mutants
US6489293B1 (en) 1992-07-28 2002-12-03 Beth Israel Deaconess Medical Center Recombinant human erythropoietin with altered biological activity
US5989538A (en) * 1995-02-15 1999-11-23 Amgen Inc. Mpl ligand analogs
US5756083A (en) * 1995-02-15 1998-05-26 Amgen Inc. Mpl ligand analogs
US5696250A (en) * 1995-02-15 1997-12-09 Amgen Inc. DNA encoding megakaryocyte growth and development factor analogs
US7399833B1 (en) 1995-02-15 2008-07-15 Elliott Steven G MGDF analogs
WO1997013783A1 (en) * 1995-10-09 1997-04-17 Pharmacia & Upjohn Company An acceptor polypeptide for an n-acetylgalactosaminyltransferase
KR100400637B1 (en) * 1996-02-06 2004-03-10 씨제이 주식회사 Biologically useful peptide
US7105484B2 (en) 1996-03-14 2006-09-12 Genentech, Inc. GDNF receptorα
US6504007B1 (en) 1996-03-14 2003-01-07 Genentech, Inc. GDNF receptor
US7691606B2 (en) 1996-03-14 2010-04-06 Genentech, Inc. Polynucleotides encoding GDNFRα
US6548653B1 (en) 1998-06-15 2003-04-15 Genzyme Transgenics Corporation Erythropoietin analog-human serum albumin fusion
US7101971B2 (en) 1998-06-15 2006-09-05 Gtc Biotherapeutics, Inc. Erythropoietin analog-human serum albumin fusion
US7304150B1 (en) 1998-10-23 2007-12-04 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
WO2000024893A3 (en) * 1998-10-23 2000-09-14 Amgen Inc Methods and compositions for the prevention and treatment of anemia
EP1813624A1 (en) * 1998-10-23 2007-08-01 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
AU774989B2 (en) * 1998-10-23 2004-07-15 Amgen, Inc. Methods and compositions for the prevention and treatment of anemia
US7973009B2 (en) 1998-10-23 2011-07-05 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
US8268588B2 (en) 1998-10-23 2012-09-18 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
WO2000024893A2 (en) * 1998-10-23 2000-05-04 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
US7977311B2 (en) 1998-10-23 2011-07-12 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
US7514072B1 (en) 1998-12-14 2009-04-07 Hannelore Ehrenreich Method for the treatment of cerebral ischaemia and use of erythropoietin or erythropoietin derivatives for the treatment of cerebral ischaemia
WO2000047741A1 (en) * 1999-02-12 2000-08-17 Amgen Inc. Glycosylated leptin compositions and related methods
US7345019B1 (en) 1999-04-13 2008-03-18 The Kenneth S. Warren Institute, Inc. Modulation of excitable tissue function by peripherally administered erythropoietin
US7309687B1 (en) 1999-04-13 2007-12-18 The Kenneth S. Warren Institute, Inc. Methods for treatment and prevention of neuromuscular and muscular conditions by peripherally administered erythropoietin
US7410941B1 (en) 1999-04-13 2008-08-12 The Kenneth S. Warren Institute, Inc. Methods for treatment of neurodegenerative conditions by peripherally administered erythropoietin
US7132280B2 (en) 1999-04-15 2006-11-07 Crucell Holland, B.V. Recombinant protein production in a human cell
US7604960B2 (en) 1999-04-15 2009-10-20 Crucell Holland B.V. Transient protein expression methods
US7833753B2 (en) 1999-04-15 2010-11-16 Crucell Holland B.V. Methods of producing erythropoietin isoforms comprising Lewis-X structures and high sialic acid content and compositions of the same
US7491532B2 (en) 1999-04-15 2009-02-17 Crucell Holland, B.V. Recombinant protein production in a human cell
US8236561B2 (en) 1999-04-15 2012-08-07 Crucell Holland B.V. Efficient production of IgA in recombinant mammalian cells
US7470523B2 (en) 1999-04-15 2008-12-30 Crucell Holland B.V. Recombinant protein production in a human cell
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
WO2000068376A1 (en) * 1999-05-07 2000-11-16 Genentech, Inc. Novel chimpanzee erythropoietin (chepo) polypeptides and nucleic acids encoding the same
JP2002543784A (en) * 1999-05-07 2002-12-24 ジェネンテック・インコーポレーテッド Novel chimpanzee erythropoietin (CHEPO) polypeptide and nucleic acid encoding the same
US6831060B2 (en) 1999-05-07 2004-12-14 Genentech, Inc. Chimpanzee erythropoietin (CHEPO) polypeptides and nucleic acids encoding the same
US6555343B1 (en) 1999-05-07 2003-04-29 Genentech Inc. Chimpanzee erythropoietin (CHEPO) polypeptides and nucleic acids encoding the same
US6583272B1 (en) * 1999-07-02 2003-06-24 Hoffmann-La Roche Inc. Erythropoietin conjugates
US7964198B2 (en) 1999-11-26 2011-06-21 Crucell Holland B.V. Production of vaccines
US7833788B2 (en) 1999-11-26 2010-11-16 Crucell Holland B.V. Production of vaccines
US7521220B2 (en) 1999-11-26 2009-04-21 Crucell Holland B.V. Production of vaccines
US7527961B2 (en) 1999-11-26 2009-05-05 Crucell Holland B.V. Production of vaccines
US7550284B2 (en) 1999-11-26 2009-06-23 Crucell Holland B.V. Production of vaccines
US7192759B1 (en) 1999-11-26 2007-03-20 Crucell Holland B.V. Production of vaccines
US7262166B2 (en) * 2000-04-07 2007-08-28 Amgen Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
US6586398B1 (en) * 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
EP2281828A2 (en) 2000-12-29 2011-02-09 The Kenneth S. Warren Institute, Inc. Compositions comprising modified erythropoietin
US6531121B2 (en) 2000-12-29 2003-03-11 The Kenneth S. Warren Institute, Inc. Protection and enhancement of erythropoietin-responsive cells, tissues and organs
US7041794B2 (en) * 2001-04-04 2006-05-09 Genodyssee Polynucleotides and polypeptides of the erythropoietin gene
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8716239B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
DE10235248B4 (en) * 2001-11-29 2004-12-23 Cheil Jedang Corp. Fusion protein with enhanced in vivo activity of erythropoietin
US7091326B2 (en) 2001-12-03 2006-08-15 Cheil Jedang Corporation Fusion protein having enhanced in vivo erythropoietin activity
US8802417B2 (en) 2001-12-07 2014-08-12 Crucell Holland B.V. Production of viruses, viral isolates and vaccines
US7504248B2 (en) 2001-12-07 2009-03-17 Crucell Holland B.V. Production of viruses viral isolates and vaccines
US8252739B2 (en) 2001-12-21 2012-08-28 Human Genome Sciences, Inc. Albumin fusion proteins
US7847079B2 (en) 2001-12-21 2010-12-07 Human Genome Sciences, Inc. Albumin fusion proteins
US8513189B2 (en) 2001-12-21 2013-08-20 Human Genome Sciences, Inc. Albumin fusion proteins
US9296809B2 (en) 2001-12-21 2016-03-29 Human Genome Sciences, Inc. Albumin fusion proteins
US8993517B2 (en) 2001-12-21 2015-03-31 Human Genome Sciences, Inc. Albumin fusion proteins
US9221896B2 (en) 2001-12-21 2015-12-29 Human Genome Sciences, Inc. Albumin fusion proteins
US8071539B2 (en) 2001-12-21 2011-12-06 Human Genome Sciences, Inc. Albumin fusion proteins
US8633153B2 (en) 2002-04-04 2014-01-21 Amgen Inc. Transthyretin variants
EP2191838A1 (en) 2002-07-26 2010-06-02 EPOPLUS GmbH &amp; Co. KG Erythropoetin for the stimulation of endothelial precursor cells
US8114839B2 (en) * 2002-09-09 2012-02-14 Hanall Biopharma Co., Ltd. Protease resistant modified erythropoietin polypeptides
US8853161B2 (en) 2003-04-09 2014-10-07 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US7291484B2 (en) 2003-05-09 2007-11-06 Crucell Holland B.V. Processes for culturing E1-immortalized cells to increase product yields
US8008043B2 (en) 2003-05-09 2011-08-30 Crucell Holland B.V. Cultures of E1-immortalized cells and processes for culturing the same to increase product yields therefrom
US7608431B2 (en) 2003-05-09 2009-10-27 Crucell Holland B.V. Fed-batch process for production of erythropoietin in human embryonic retina cells that express adenovirus E1A
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
EP2156843A2 (en) 2004-01-23 2010-02-24 EPOPLUS GmbH &amp; Co. KG Use of low-dose erythropoietin for the treatment of acute or chronic renal insufficiency and for the treatment of wounds
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
US10874714B2 (en) 2004-10-29 2020-12-29 89Bio Ltd. Method of treating fibroblast growth factor 21 (FGF-21) deficiency
EP2402450A1 (en) 2004-12-30 2012-01-04 Crucell Holland B.V. Methods to obtain recombinant proteins with decreased LacdiNac structures
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
WO2006120030A1 (en) 2005-05-13 2006-11-16 Charite Universitätsmedizin - Berlin Erythropoietin variants
US9988427B2 (en) 2005-05-13 2018-06-05 Charite Universitaetsmedizen-Berlin Erythropoietin variants
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
EP1957630B1 (en) 2005-12-08 2016-02-17 Amgen Inc. Improved production of glycoproteins using manganese
EP2495308A1 (en) 2005-12-08 2012-09-05 Amgen Inc. Improved production of glycoproteins using manganese
US8460896B2 (en) 2005-12-08 2013-06-11 Amgen Inc. Host cells and culture methods
EP3026109A1 (en) 2005-12-08 2016-06-01 Amgen Inc. Improved production of glycoproteins using manganese
US7888101B2 (en) 2005-12-08 2011-02-15 Amgen Inc. Host cells comprising alpha 1,2 mannosidase and culture methods thereof
US7972810B2 (en) 2005-12-08 2011-07-05 Amgen Inc. Production of glycoproteins using manganese
EP4155385A1 (en) 2005-12-08 2023-03-29 Amgen Inc. Improved production of glycoproteins using manganese
US8617878B2 (en) 2005-12-08 2013-12-31 Amgen Inc. Production of glycoproteins using manganese
US9273110B2 (en) 2005-12-08 2016-03-01 Amgen Inc. Production of glycoproteins using manganese
US8680248B2 (en) 2005-12-08 2014-03-25 Amgen Inc. Host cells comprising alpha 1,2 mannosidase and culture methods thereof
EP1957630B2 (en) 2005-12-08 2023-03-29 Amgen Inc. Improved production of glycoproteins using manganese
US8247210B2 (en) 2005-12-08 2012-08-21 Amgen Inc. Host cells comprising alpha 1,2 mannosidase and culture methods thereof
EP3026109B1 (en) * 2005-12-08 2022-03-09 Amgen Inc. Improved production of glycoproteins using manganese
US9254312B2 (en) 2006-01-27 2016-02-09 Hannelore Ehrenreich Method for the treatment and/or prophylaxis of multiple sclerosis, and use of erythropoietin for the manufacture of a medicament for the intermittent treatment and/or intermittent prophylaxis of multiple sclerosis
EP2319530A1 (en) 2006-01-27 2011-05-11 Hannelore Ehrenreich Method for treating and/or preventing multiple sclerosis, and use of erythropoietin in the production of a drug for intermittent treatment and/or intermittent prevention of multiple sclerosis
WO2007108505A1 (en) 2006-03-22 2007-09-27 Chugai Seiyaku Kabushiki Kaisha Erythropoietin solution preparation
US8546329B2 (en) 2006-03-22 2013-10-01 Chugai Seiyaku Kabushiki Kaisha Erythropoietin solution preparation
US7851438B2 (en) 2006-05-19 2010-12-14 GlycoFi, Incorporated Erythropoietin compositions
US8137954B2 (en) 2006-05-19 2012-03-20 Glycofi, Inc. Erythropoietin compositions
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
EP2498096A1 (en) 2006-07-21 2012-09-12 Amgen Inc. Method of detecting and/or measuring hepcidin in a sample
WO2008023725A1 (en) 2006-08-22 2008-02-28 Chugai Seiyaku Kabushiki Kaisha Preventive and/or therapeutic agent for peripheral neuropathy
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US8895303B2 (en) 2006-11-13 2014-11-25 Charite-Universitatsmedizin Berlin Method of cell culture and method of treatment comprising a vEPO protein variant
WO2008111503A1 (en) 2007-03-09 2008-09-18 National University Corporation Obihiro University Of Agriculture And Veterinary Medicine Protective agent for transplanted organ
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
US9180165B2 (en) 2007-07-19 2015-11-10 Hannelore Ehrenreich Use of EPO receptor activation or stimulation for the improvement of the EDSS score in patients with multiple sclerosis
US9320797B2 (en) 2007-09-27 2016-04-26 Amgen Inc. Pharmaceutical formulations
US10653781B2 (en) 2007-09-27 2020-05-19 Amgen Inc. Pharmaceutical formulations
EP3381445A2 (en) 2007-11-15 2018-10-03 Amgen Inc. Aqueous formulation of antibody stablised by antioxidants for parenteral administration
US9688759B2 (en) 2008-01-25 2017-06-27 Amgen, Inc. Ferroportin antibodies and methods of use
WO2009094551A1 (en) 2008-01-25 2009-07-30 Amgen Inc. Ferroportin antibodies and methods of use
EP2574628A1 (en) 2008-01-25 2013-04-03 Amgen Inc. Ferroportin antibodies and methods of use
US9175078B2 (en) 2008-01-25 2015-11-03 Amgen Inc. Ferroportin antibodies and methods of use
EP2803675A2 (en) 2008-01-25 2014-11-19 Amgen, Inc Ferroportin antibodies and methods of use
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
EP2620448A1 (en) 2008-05-01 2013-07-31 Amgen Inc. Anti-hepcidin antibodies and methods of use
EP2816059A1 (en) 2008-05-01 2014-12-24 Amgen, Inc Anti-hepcidin antibodies and methods of use
EP3693014A1 (en) 2008-11-13 2020-08-12 The General Hospital Corporation Methods and compositions for regulating iron homeostasis by modulation bmp-6
WO2010056981A2 (en) 2008-11-13 2010-05-20 Massachusetts General Hospital Methods and compositions for regulating iron homeostasis by modulation bmp-6
US9517247B2 (en) 2009-04-27 2016-12-13 Cytonet Gmbh & Co. Kg Encapsulated liver cell composition
US8535923B2 (en) 2009-04-27 2013-09-17 Cytonet Gmbh & Co. Kg Encapsulated liver cell composition
WO2011011674A2 (en) 2009-07-24 2011-01-27 Dr. Reddy's Laboratories Ltd. Production of erythropoiesis stimulating protein using metal ions
EP3009447A1 (en) 2009-07-24 2016-04-20 Dr. Reddy's Laboratories, Ltd. Production of erythropoiesis stimulating protein using metal ions
WO2011050333A1 (en) 2009-10-23 2011-04-28 Amgen Inc. Vial adapter and system
WO2011156373A1 (en) 2010-06-07 2011-12-15 Amgen Inc. Drug delivery device
WO2012135315A1 (en) 2011-03-31 2012-10-04 Amgen Inc. Vial adapter and system
EP4074355A1 (en) 2011-04-20 2022-10-19 Amgen Inc. Autoinjector apparatus
WO2013055873A1 (en) 2011-10-14 2013-04-18 Amgen Inc. Injector and method of assembly
EP3335747A1 (en) 2011-10-14 2018-06-20 Amgen Inc. Injector and method of assembly
EP3269413A1 (en) 2011-10-14 2018-01-17 Amgen, Inc Injector and method of assembly
EP3744371A1 (en) 2011-10-14 2020-12-02 Amgen, Inc Injector and method of assembly
EP3045188A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3045190A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3045187A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
EP3045189A1 (en) 2011-10-14 2016-07-20 Amgen, Inc Injector and method of assembly
CN102788720A (en) * 2012-06-05 2012-11-21 西北大学 Membrane assisted separation of glycoprotein all N-linked carbohydrate chain and identification method thereof
US11458247B2 (en) 2012-11-21 2022-10-04 Amgen Inc. Drug delivery device
US11439745B2 (en) 2012-11-21 2022-09-13 Amgen Inc. Drug delivery device
EP3072548A1 (en) 2012-11-21 2016-09-28 Amgen, Inc Drug delivery device
EP3656426A1 (en) 2012-11-21 2020-05-27 Amgen, Inc Drug delivery device
US10682474B2 (en) 2012-11-21 2020-06-16 Amgen Inc. Drug delivery device
EP3081249A1 (en) 2012-11-21 2016-10-19 Amgen, Inc Drug delivery device
EP4234694A2 (en) 2012-11-21 2023-08-30 Amgen Inc. Drug delivery device
WO2014081780A1 (en) 2012-11-21 2014-05-30 Amgen Inc. Drug delivery device
US11344681B2 (en) 2012-11-21 2022-05-31 Amgen Inc. Drug delivery device
US10428129B2 (en) 2012-12-18 2019-10-01 Ubi Pharma Inc. Recombinant protein
US10239941B2 (en) 2013-03-15 2019-03-26 Intrinsic Lifesciences Llc Anti-hepcidin antibodies and uses thereof
US9803011B2 (en) 2013-03-15 2017-10-31 Intrinsic Lifesciences Llc Anti-hepcidin antibodies and uses thereof
US9657098B2 (en) 2013-03-15 2017-05-23 Intrinsic Lifesciences, Llc Anti-hepcidin antibodies and uses thereof
WO2014144096A1 (en) 2013-03-15 2014-09-18 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
WO2014149357A1 (en) 2013-03-22 2014-09-25 Amgen Inc. Injector and method of assembly
EP3831427A1 (en) 2013-03-22 2021-06-09 Amgen Inc. Injector and method of assembly
WO2015061386A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Injector and method of assembly
EP3789064A1 (en) 2013-10-24 2021-03-10 Amgen, Inc Injector and method of assembly
WO2015061389A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Drug delivery system with temperature-sensitive control
EP3421066A1 (en) 2013-10-24 2019-01-02 Amgen, Inc Injector and method of assembly
EP3501575A1 (en) 2013-10-24 2019-06-26 Amgen, Inc Drug delivery system with temperature-sensitive-control
US10723775B2 (en) 2013-11-29 2020-07-28 Cj Healthcare Corporation Method for purifying darbepoetin alfa
WO2015080509A1 (en) 2013-11-29 2015-06-04 씨제이헬스케어 주식회사 Method for purifying darbepoetin alfa
WO2015119906A1 (en) 2014-02-05 2015-08-13 Amgen Inc. Drug delivery system with electromagnetic field generator
WO2015171777A1 (en) 2014-05-07 2015-11-12 Amgen Inc. Autoinjector with shock reducing elements
EP3785749A1 (en) 2014-05-07 2021-03-03 Amgen Inc. Autoinjector with shock reducing elements
WO2015187797A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Controllable drug delivery system and method of use
WO2015187793A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Drug delivery system and method of use
WO2015187799A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Systems and methods for remotely processing data collected by a drug delivery device
US11213624B2 (en) 2014-06-03 2022-01-04 Amgen Inc. Controllable drug delivery system and method of use
US11738146B2 (en) 2014-06-03 2023-08-29 Amgen Inc. Drug delivery system and method of use
EP4036924A1 (en) 2014-06-03 2022-08-03 Amgen, Inc Devices and methods for assisting a user of a drug delivery device
US10323088B2 (en) 2014-09-22 2019-06-18 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof
WO2016049036A1 (en) 2014-09-22 2016-03-31 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof
EP3943135A2 (en) 2014-10-14 2022-01-26 Amgen Inc. Drug injection device with visual and audible indicators
WO2016061220A2 (en) 2014-10-14 2016-04-21 Amgen Inc. Drug injection device with visual and audio indicators
EP3848072A1 (en) 2014-12-19 2021-07-14 Amgen Inc. Drug delivery device with proximity sensor
WO2016100055A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with live button or user interface field
EP3689394A1 (en) 2014-12-19 2020-08-05 Amgen Inc. Drug delivery device with live button or user interface field
WO2016100781A1 (en) 2014-12-19 2016-06-23 Amgen Inc. Drug delivery device with proximity sensor
US10765801B2 (en) 2014-12-19 2020-09-08 Amgen Inc. Drug delivery device with proximity sensor
US11357916B2 (en) 2014-12-19 2022-06-14 Amgen Inc. Drug delivery device with live button or user interface field
US10799630B2 (en) 2014-12-19 2020-10-13 Amgen Inc. Drug delivery device with proximity sensor
US11944794B2 (en) 2014-12-19 2024-04-02 Amgen Inc. Drug delivery device with proximity sensor
EP3556411A1 (en) 2015-02-17 2019-10-23 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
EP3981450A1 (en) 2015-02-27 2022-04-13 Amgen, Inc Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
WO2017100501A1 (en) 2015-12-09 2017-06-15 Amgen Inc. Auto-injector with signaling cap
WO2017120178A1 (en) 2016-01-06 2017-07-13 Amgen Inc. Auto-injector with signaling electronics
WO2017160799A1 (en) 2016-03-15 2017-09-21 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
EP4035711A1 (en) 2016-03-15 2022-08-03 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
EP3721922A1 (en) 2016-03-15 2020-10-14 Amgen Inc. Reducing probability of glass breakage in drug delivery devices
WO2017189089A1 (en) 2016-04-29 2017-11-02 Amgen Inc. Drug delivery device with messaging label
WO2017192287A1 (en) 2016-05-02 2017-11-09 Amgen Inc. Syringe adapter and guide for filling an on-body injector
WO2017197222A1 (en) 2016-05-13 2017-11-16 Amgen Inc. Vial sleeve assembly
WO2017200989A1 (en) 2016-05-16 2017-11-23 Amgen Inc. Data encryption in medical devices with limited computational capability
WO2017209899A1 (en) 2016-06-03 2017-12-07 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
WO2018004842A1 (en) 2016-07-01 2018-01-04 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
WO2018034784A1 (en) 2016-08-17 2018-02-22 Amgen Inc. Drug delivery device with placement detection
WO2018081234A1 (en) 2016-10-25 2018-05-03 Amgen Inc. On-body injector
WO2018136398A1 (en) 2017-01-17 2018-07-26 Amgen Inc. Injection devices and related methods of use and assembly
WO2018151890A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Drug delivery device with sterile fluid flowpath and related method of assembly
WO2018152073A1 (en) 2017-02-17 2018-08-23 Amgen Inc. Insertion mechanism for drug delivery device
WO2018165143A1 (en) 2017-03-06 2018-09-13 Amgen Inc. Drug delivery device with activation prevention feature
WO2018164829A1 (en) 2017-03-07 2018-09-13 Amgen Inc. Needle insertion by overpressure
WO2018165499A1 (en) 2017-03-09 2018-09-13 Amgen Inc. Insertion mechanism for drug delivery device
WO2018172219A1 (en) 2017-03-20 2018-09-27 F. Hoffmann-La Roche Ag Method for in vitro glycoengineering of an erythropoiesis stimulating protein
EP4241807A2 (en) 2017-03-28 2023-09-13 Amgen Inc. Plunger rod and syringe assembly system and method
WO2018183039A1 (en) 2017-03-28 2018-10-04 Amgen Inc. Plunger rod and syringe assembly system and method
WO2018226565A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Torque driven drug delivery device
WO2018226515A1 (en) 2017-06-08 2018-12-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
WO2018236619A1 (en) 2017-06-22 2018-12-27 Amgen Inc. Device activation impact/shock reduction
WO2018237225A1 (en) 2017-06-23 2018-12-27 Amgen Inc. Electronic drug delivery device comprising a cap activated by a switch assembly
WO2019014014A1 (en) 2017-07-14 2019-01-17 Amgen Inc. Needle insertion-retraction system having dual torsion spring system
EP4292576A2 (en) 2017-07-21 2023-12-20 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
WO2019018169A1 (en) 2017-07-21 2019-01-24 Amgen Inc. Gas permeable sealing member for drug container and methods of assembly
WO2019022951A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with gear module and related method of assembly
WO2019022950A1 (en) 2017-07-25 2019-01-31 Amgen Inc. Drug delivery device with container access system and related method of assembly
EP4085942A1 (en) 2017-07-25 2022-11-09 Amgen Inc. Drug delivery device with gear module and related method of assembly
WO2019032482A2 (en) 2017-08-09 2019-02-14 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
WO2019036181A1 (en) 2017-08-18 2019-02-21 Amgen Inc. Wearable injector with sterile adhesive patch
WO2019040548A1 (en) 2017-08-22 2019-02-28 Amgen Inc. Needle insertion mechanism for drug delivery device
WO2019070472A1 (en) 2017-10-04 2019-04-11 Amgen Inc. Flow adapter for drug delivery device
EP4257164A2 (en) 2017-10-06 2023-10-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
WO2019070552A1 (en) 2017-10-06 2019-04-11 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
WO2019074579A1 (en) 2017-10-09 2019-04-18 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
WO2019090079A1 (en) 2017-11-03 2019-05-09 Amgen Inc. System and approaches for sterilizing a drug delivery device
WO2019090086A1 (en) 2017-11-03 2019-05-09 Amgen Inc. Systems and approaches for sterilizing a drug delivery device
WO2019090303A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Fill-finish assemblies and related methods
WO2019089178A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Drug delivery device with placement and flow sensing
WO2019094138A1 (en) 2017-11-10 2019-05-16 Amgen Inc. Plungers for drug delivery devices
WO2019099322A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Autoinjector with stall and end point detection
WO2019099324A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Door latch mechanism for drug delivery device
WO2019231582A1 (en) 2018-05-30 2019-12-05 Amgen Inc. Thermal spring release mechanism for a drug delivery device
WO2019231618A1 (en) 2018-06-01 2019-12-05 Amgen Inc. Modular fluid path assemblies for drug delivery devices
WO2020023444A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
WO2020023451A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Delivery devices for administering drugs
WO2020023220A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with tacky skin attachment portion and related method of preparation
WO2020028009A1 (en) 2018-07-31 2020-02-06 Amgen Inc. Fluid path assembly for a drug delivery device
WO2020068623A1 (en) 2018-09-24 2020-04-02 Amgen Inc. Interventional dosing systems and methods
WO2020065576A1 (en) 2018-09-27 2020-04-02 Universidad Nacional Del Litoral Modified human erythropoietin
WO2020068476A1 (en) 2018-09-28 2020-04-02 Amgen Inc. Muscle wire escapement activation assembly for a drug delivery device
WO2020072577A1 (en) 2018-10-02 2020-04-09 Amgen Inc. Injection systems for drug delivery with internal force transmission
WO2020072846A1 (en) 2018-10-05 2020-04-09 Amgen Inc. Drug delivery device having dose indicator
WO2020081480A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Platform assembly process for drug delivery device
WO2020081479A1 (en) 2018-10-15 2020-04-23 Amgen Inc. Drug delivery device having damping mechanism
WO2020091956A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020091981A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
WO2020092056A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial needle retraction
WO2020219482A1 (en) 2019-04-24 2020-10-29 Amgen Inc. Syringe sterilization verification assemblies and methods
WO2021041067A2 (en) 2019-08-23 2021-03-04 Amgen Inc. Drug delivery device with configurable needle shield engagement components and related methods
WO2022246055A1 (en) 2021-05-21 2022-11-24 Amgen Inc. Method of optimizing a filling recipe for a drug container
WO2023209074A1 (en) 2022-04-28 2023-11-02 Institut National de la Santé et de la Recherche Médicale Methods of restoring erythropoiesis in patients suffering from a sf3b1 mutant myelodysplastic syndrome by correcting coasy mis-splicing

Also Published As

Publication number Publication date
NL300075I2 (en) 2002-04-02
DE69404401T3 (en) 2005-11-24
JPH11155584A (en) 1999-06-15
DE10199059I1 (en) 2002-02-07
JP2938572B2 (en) 1999-08-23
CZ91795A3 (en) 1996-03-13
CA2147124C (en) 2002-11-05
JP2004331671A (en) 2004-11-25
DE10199059I2 (en) 2007-11-08
FI117136B (en) 2006-06-30
HU220334B (en) 2001-12-28
KR100328769B1 (en) 2002-08-08
ES2105442T3 (en) 1997-10-16
ATE155796T1 (en) 1997-08-15
CA2147124A1 (en) 1995-02-23
DK0640619T3 (en) 1998-02-02
NO2007008I1 (en) 2007-07-23
DK0640619T4 (en) 2005-04-18
SI0640619T1 (en) 1998-02-28
FI951792A (en) 1995-05-18
AU7632794A (en) 1995-03-14
NZ273134A (en) 1997-11-24
SK50295A3 (en) 1995-08-09
NO951445L (en) 1995-06-16
NO951445D0 (en) 1995-04-12
IL110669A (en) 2008-11-26
JP3664590B2 (en) 2005-06-29
SI0640619T2 (en) 2005-06-30
CN1057534C (en) 2000-10-18
JP3664723B2 (en) 2005-06-29
LV10972B (en) 1996-10-20
KR960701994A (en) 1996-03-28
JPH08506023A (en) 1996-07-02
HU9501435D0 (en) 1995-06-28
FI951792A0 (en) 1995-04-13
NO2007008I2 (en) 2011-02-21
RU95115239A (en) 1997-03-27
CN1105030A (en) 1995-07-12
EP0640619B2 (en) 2005-03-23
HUT72849A (en) 1996-05-28
AU677097B2 (en) 1997-04-10
IL136188A0 (en) 2001-07-24
NL300075I1 (en) 2002-03-01
IL192290A0 (en) 2008-12-29
DE69404401T2 (en) 1998-02-19
IL110669A0 (en) 1994-11-11
UA49793C2 (en) 2002-10-15
NO323104B1 (en) 2007-01-02
HK1001589A1 (en) 1998-06-26
ES2105442T5 (en) 2005-07-16
SK282003B6 (en) 2001-10-08
EP0640619B1 (en) 1997-07-23
IL136189A0 (en) 2001-07-24
RU2159814C2 (en) 2000-11-27
CZ291229B6 (en) 2003-01-15
ZA946122B (en) 1995-03-20
LV10972A (en) 1995-12-20
CZ291342B6 (en) 2003-02-12
EP0640619A1 (en) 1995-03-01
DE69404401D1 (en) 1997-09-04
GR3024815T3 (en) 1998-01-30
LU90850I2 (en) 2002-01-07
CZ291343B6 (en) 2003-02-12

Similar Documents

Publication Publication Date Title
AU677097B2 (en) Erythropoietin analogs
US7217689B1 (en) Glycosylation analogs of erythropoietin
US5856298A (en) Erythropoietin isoforms
AU646822B2 (en) Erythropoietin isoforms
JPH05103675A (en) Production of human nerve growth factor 2
IE83805B1 (en) Erythropoietin analogs

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BB BG BR BY CA CH CN CZ DE DK ES FI GB HU JP KP KR KZ LK LT LU LV MG MN MW NL NO NZ PL PT RO RU SD SE SK UA UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: PV1995-917

Country of ref document: CZ

WWE Wipo information: entry into national phase

Ref document number: 951792

Country of ref document: FI

Ref document number: 2147124

Country of ref document: CA

Ref document number: 273134

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1019950701453

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 50295

Country of ref document: SK

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
WWP Wipo information: published in national office

Ref document number: PV2001-463

Country of ref document: CZ

Ref document number: PV1995-917

Country of ref document: CZ

Ref document number: PV2001-462

Country of ref document: CZ

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

EX32 Extension under rule 32 effected after completion of technical preparation for international publication

Free format text: TM

WWE Wipo information: entry into national phase

Ref document number: PV2001-462

Country of ref document: CZ

Ref document number: PV2001-463

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: PV1995-917

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: PV2001-463

Country of ref document: CZ

Ref document number: PV2001-462

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 951792

Country of ref document: FI